TW201200525A - MYBL2 peptides and vaccines containing the same - Google Patents

MYBL2 peptides and vaccines containing the same Download PDF

Info

Publication number
TW201200525A
TW201200525A TW099141433A TW99141433A TW201200525A TW 201200525 A TW201200525 A TW 201200525A TW 099141433 A TW099141433 A TW 099141433A TW 99141433 A TW99141433 A TW 99141433A TW 201200525 A TW201200525 A TW 201200525A
Authority
TW
Taiwan
Prior art keywords
peptide
antigen
cancer
cytotoxic
present
Prior art date
Application number
TW099141433A
Other languages
Chinese (zh)
Inventor
Yusuke Nakamura
Takuya Tsunoda
Ryuji Ohsawa
Sachiko Yoshimura
Tomohisa Watanabe
Original Assignee
Oncotherapy Science Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oncotherapy Science Inc filed Critical Oncotherapy Science Inc
Publication of TW201200525A publication Critical patent/TW201200525A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

Peptide vaccines against cancer are described herein. In particular, epitope peptides derived from the MYBL2 gene that bind to HLA antigen and have cytotoxic T lymphocyte (CTL) inducibility, more particularly peptides having the amino acid sequence of SEQ ID NO: 5 and fragments thereof, are provided. The present invention further extends to peptides that include one, two, or several amino acid insertions, substitutions or additions to the aforementioned peptides or fragments, provided they retain cytotoxic T cell inducibility. Also provided as nucleic acids encoding any of the aforementioned peptides, antigen-presenting cells and isolated CTLs that target such peptides, and pharmaceutical agents and compositions including any of the aforementioned peptides, nucleic acids, and APCs as active ingredients. The components of the present invention have particular utility in connection with the treatment and/or prophylaxis (i.e., prevention) of cancers (tumors), and/or the prevention of a postoperative recurrence thereof.

Description

201200525 化學式 五本案右有化學式時,請揭示最能顯示發明特 無。 六、發明說明: 【發明所屬之技術領域】 本發明係關於生物科學領域’更特別對於癌症治療領 域。特別是,本發明係關於新穎之胜肽,纟當作癌症疫苗 及治療與避免腫瘤之藥物為非常有效。 【先前技術】 已證實CD8陽性細胞毒殺性τ淋巴球辨認來自建造於 主要組織相容性抗原複合體(ina j〇r hist〇c〇mpatibi丨丨衿 complex’ MHC) class I分子上之腫瘤相關抗原(1;_〇卜 associated antigens,TAAs)的抗原決定位胜肽,且之後 殺死腫瘤細胞。自從發現黑色素瘤抗原(melan〇maantigen MAGE)家族為腫瘤相關抗原之第一個例子,藉由免疫方法, 已發現許多其他腫瘤相關抗原(NPL 1, Boon T, Int J Cancer 1 993 May 8, 54(2): 1 77-80; NPL 2, Boon T & van der Bruggen P, J Exp Med 1 996 Mar 1,183(3): 725-9) ’ 且這些腫瘤相關抗原的一些目前在臨床發展的過程中作為 免疫治療標的。 能誘導有效且專一之抗腫瘤免疫反應的新腫瘤相關抗 原的辨認成為於多種形式癌症中之胜肽疫苗接種策略 (vaccination strategies)之更進一步發展與臨床研究的 201200525 根據為正進行著(NPL 3, Harris CC,J Natl Cancer Inst 1996 Oct 16, 88(20): 1442-55; NPL 4, Butterfield LH et al., Cancer Res 1999 Jul 1, 59(13): 3134-42; NPL 5, Vissers JL et al. , Cancer Res 1999 Nov 1, 59(21): 5554-9; NPL 6, van der Burg SH et al. , J Immunol 1996 May 1, 1 56(9): 3308-14; NPL 7, Tanaka F et al. , Cancer Res 1 997 Oct 1 5, 57(20): 4465-8; NPL 8, Fujie T ei a;., Int J Cancer 1 999 Jan 18, 80(2): 1 69-72; NPL 9, Kikuchi M et al., Int J Cancer 1 999 May 5, 81 (3): 459-66; NPL 10, Oiso M et al., Int J Cancer 1999 May 5, 81(3): 387-94)。迄今已報導許多使用這些腫瘤相關抗原衍生胜肽 的臨床試驗。不幸地,許多目前癌症疫苗試驗已顯示一低 的客觀反應率(objective response rate) (NPL 11,Be 11 i F et al., J Clin Oncol 2002 Oct 15, 20(20): 4169-80; NPL 12, Coulie PG et al. , Immunol Rev 2002 Oct, 188: 33-42; NPL 13, Rosenberg SA et al. , Nat Med 2004 Sep, 1 0 ( 9 ): 909-1 5)。因此,仍然需要新穎之腫瘤相關抗原作 為免疫治療標的。 以c-myb原致癌基因探針藉由cDNA資料庫的篩選,已 鑑定出 MYBL2 ( GenBank Accession No: NM_002466,編碼 出序列辨識:6的序列辨識號:5 ),類v-myb骨髓胚細胞 過多症病毒致癌同系物2 (v-myb myeloblastosis viral oncogene homolog (avian)-like 2),為轉錄因子基因之 MYB 家族的一員(NPL 14,Nomura N et 3人,Nucleic Acids 4 201200525201200525 Chemical Formula 5 When there is a chemical formula on the right, please reveal the best indication of the invention. 6. Description of the Invention: TECHNICAL FIELD OF THE INVENTION The present invention relates to the field of biological sciences, and more particularly to the field of cancer treatment. In particular, the present invention relates to a novel peptide which is very effective as a cancer vaccine and a drug for treating and avoiding tumors. [Prior Art] It has been confirmed that CD8-positive cells are toxic to the tumor-associated antigen-complex (ina j〇r hist〇c〇mpatibi丨丨衿complex' MHC) class I tumor-associated tumor-associated The epitopes of the antigen (1; _ associated antigens, TAAs) are peptides, and then the tumor cells are killed. Since the discovery of the melanoma antigen family (melan〇maantigen MAGE) family as the first example of tumor-associated antigens, many other tumor-associated antigens have been discovered by immunological methods (NPL 1, Boon T, Int J Cancer 1 993 May 8, 54 (2): 1 77-80; NPL 2, Boon T & van der Bruggen P, J Exp Med 1 996 Mar 1,183(3): 725-9) ' and some of these tumor-associated antigens are currently in clinical development The process is labeled as an immunotherapy. The identification of new tumor-associated antigens that induce an effective and specific anti-tumor immune response has become a further development and clinical study of peptide vaccination strategies in various forms of cancer. 201200525 Based on ongoing (NPL 3 , Harris CC, J Natl Cancer Inst 1996 Oct 16, 88(20): 1442-55; NPL 4, Butterfield LH et al., Cancer Res 1999 Jul 1, 59(13): 3134-42; NPL 5, Vissers JL Et al. , Cancer Res 1999 Nov 1, 59(21): 5554-9; NPL 6, van der Burg SH et al. , J Immunol 1996 May 1, 1 56(9): 3308-14; NPL 7, Tanaka F et al. , Cancer Res 1 997 Oct 1 5, 57(20): 4465-8; NPL 8, Fujie T ei a;., Int J Cancer 1 999 Jan 18, 80(2): 1 69-72; NPL 9, Kikuchi M et al., Int J Cancer 1 999 May 5, 81 (3): 459-66; NPL 10, Oiso M et al., Int J Cancer 1999 May 5, 81(3): 387-94 ). Many clinical trials using these tumor-associated antigen-derived peptides have been reported to date. Unfortunately, many current cancer vaccine trials have shown a low objective response rate (NPL 11, Be 11 i F et al., J Clin Oncol 2002 Oct 15, 20(20): 4169-80; NPL 12, Coulie PG et al., Immunol Rev 2002 Oct, 188: 33-42; NPL 13, Rosenberg SA et al., Nat Med 2004 Sep, 1 0 (9): 909-1 5). Therefore, novel tumor-associated antigens are still required as immunotherapeutic targets. MYBL2 (GenBank Accession No: NM_002466, sequence identification: 6 sequence identification number: 5) has been identified by c-myb proto-oncogene probe by cDNA library, and there are too many v-myb-like myeloid cells. V-myb myeloblastosis viral oncogene homolog (avian)-like 2), a member of the MYB family of transcription factor genes (NPL 14, Nomura N et 3, Nucleic Acids 4 201200525)

VV

Res· 1988 Dec 9,1 6(23): 1 1 075 - 1 1089) °MYBL2 已知 為控制細胞週期進展’與藉由CDK2-細胞週期素A(CDK2-cyclin A)與CDK2-細胞週期素E(CDK2-cyclin E)複合物之 細胞週期素驅動磷酸化(NPL 15,Robinson C ei ai., Oncogene 1996 May 2; 12(9):1855-64, NPL 16, Lane et al., Oncogene 1997 May 22; 14(20):2445-53, NPL 17,Res· 1988 Dec 9,1 6(23): 1 1 075 - 1 1089) °MYBL2 is known to control cell cycle progression' and by CDK2-cyclin A (CDK2-cyclin A) and CDK2-cyclin Cyclin-driven phosphorylation of the E(CDK2-cyclin E) complex (NPL 15, Robinson C ei ai., Oncogene 1996 May 2; 12(9): 1855-64, NPL 16, Lane et al., Oncogene 1997 May 22; 14(20): 2445-53, NPL 17,

Sala Proc Natl Acad Sci 1997 Jan 21; 94(2): 532 - 536, NPL 18, Johnson K et al., J Biol Chem 1999Sala Proc Natl Acad Sci 1997 Jan 21; 94(2): 532 - 536, NPL 18, Johnson K et al., J Biol Chem 1999

Dec 17;274(51) :36741-9)。近來報導已顯示 Mip/LIN-9 控 制MYBL2的表現且兩者蛋白質經由s與M相細胞週期素的 控制在細胞週期進展的促進中扮演一關鍵的角色(NPL 1 9,Dec 17; 274(51): 36741-9). It has recently been reported that Mip/LIN-9 controls the performance of MYBL2 and that both proteins play a key role in the promotion of cell cycle progression through the control of s and M-phase cyclins (NPL 199).

Pilkinton U et a 1. , J Biol Chem 20 07 Jan 5 ; 282(1 ) : 168-75)。此外’藉由使用含有23, 040個基因之基因體寬度CDNA 微陣列之基因表現輪廓的分析,已鑑定出MYBL2為在許多 癌症中’包括,例如睪丸腫瘤、胰臟癌、膀胱癌、非小細 胞肺癌、小細胞肺癌與食道癌被向上調控的一新穎分子 (PTL 1,US 60/937,61 6,PTL 2,W0 2004/031410)。MYBL2 被視為是一新穎的致癌抗原(oncoant igen),且來自MYBL2 的抗原決定位胜肽可被應用至具有數種形式腫瘤之病患的 癌症免疫治療。 【引用文獻】 專利文獻(Patent Literature) [PTL 1] US 60/937, 616 [PTL 2] WO 2004/031410 201200525 非專利文獻(Non Patent Literature) [NPL 1] Boon T, Int J Cancer 1993 May 8, 54(2): 177-80 [NPL 2] Boon T &amp; van der Bruggen P, J Exp Med 1996 Mar 1, 183(3): 725-9 [NPL 3] Harris CC, J Natl Cancer Inst 1 996 Oct 16, 88(20): 1442-55 [NPL 4] Butterfield LH et al. , Cancer Res 1 999 Jul 1, 59(13): 3134-42 [NPL 5] Vissers JL et al., Cancer Res 1999 Nov 1, 59(21): 5554-9 [NPL 6] van der Burg SH et al. , J Immunol 1 996 May 1, 156(9): 3308-14 [NPL 7] Tanaka F et al., Cancer Res 1 997 Oct 15, 57(20): 4465-8 [NPL 8] Fujie T et al., Int J Cancer 1 999 Jan 18, 80(2): 169-72 [NPL 9] Kikuchi M et al., Int J Cancer 1999 May 5, 81(3): 459-66 [NPL 10] Oiso M et al., Int J Cancer 1 999 May 5, 81(3): 387-94 [NPL 11] Belli F et al., J Clin Oncol 2002 Oct 15, 20(20): 4169-80 [NPL 12] Coulie PG et al., Immunol Rev 2002 Oct, ⑤ 201200525 188: 33-42 [NPL 13] Rosenberg SA et al., Nat Med 2004 Sep, 10(9): 909-15 [NPL 14] Nomura N et al., Nucleic Acids Res. 1988 Dec 9, 1 6(23): 1 1 075 - 1 1 089 [NPL 15] Robinson C et al., Oncogene 1 996 May 2; 12(9):1855-64 [NPL 16] Lane et al. , Oncogene 1997 May 22; 14(20): 2445-53 [NPL 17] Sala et al., Proc Natl Acad Sci 1 997 Jan 21; 94(2): 532 - 536 [NPL 18] Johnson K et al., J Biol Chem 1999 Dec 17;274(51):36741-9 [NPL 19] Pilkinton M et al., J Biol Chem 2007 Jan 5:282(1):168-75 【發明内容】 本發明部分基於發現免疫治療之適合的標的。由於腫 瘤相關抗原(tumor-associated antigens, TAAs)有時被免 疫系統感知為“自身”且因此常不具有免疫抗原性 (immunogenicity),所以適合標的的發現極度重要。如上 所提到,已確認MYBL2 (由GenBank獲得編號關_002466 (序列辨識號:5 )之基因所編碼出的序列辨識號:6 )在 癌症’例如睪丸腫瘤、胰臟癌、膀胱癌 '非小細胞肺癌、 201200525 小細胞肺癌與食道癌之組織中為向上調控。因此,MYBL2 為一癌症/腫瘤免疫治療之候選標的。 本發明至少部份基於具有誘導專一於MYBL2之細胞毒 殺性T淋巴球之能力的MYBL2之特定抗原決定位胜肽的確 認。詳細如下所討論,使用與Hu (人類白血球組織抗原) -A2結合之來自MYBL2的候選胜肽來刺激自健康提供者獲 得之周邊血液單核球細胞(peripheral M〇C)d mc)nc)nuclea]_ cel Is, PBMCs)。之後建立具有抗(against)經各候選胜肽 脈衝(pulsed)之HLA-A2陽性之目標細胞的專一細胞毒性 的細胞毒殺性T淋巴球。因此’這些結果證明這些胜狀為 HLA-A2限制的抗原決定位胜肽’其可誘導強而專一之抗表 現MYBL2之細胞的免疫反應。因此,這些結果顯示Μγβ[2 為強效致免疫性且其抗原決定位為腫瘤免疫治療之有效目 標。 因此,本發明一目的為提供與HLA抗原結合之經分離 的胜肽’其為MYBL2 (序列辨識號:6)或其片段。這些胜 狀被預期具有細胞毒殺性T淋巴球誘發能力且因此可被用 於&lt;?义h叩誘導細胞毒殺性τ淋巴球或可被投予至一個體 以誘導抗癌症的免疫反應,癌症的例子包括,但不限於睪 丸腫瘤、騰臟癌、膀脱癌、非小細胞肺癌、小細胞肺癌與 食道癌。較佳為’這些胜肽為九胜肽或十胜肽,且更佳為 具有擇自序列辨識號:2至4中之胺基酸序列’特別是顯 不於此之序列辨識號:3之胺基酸序列的九胜肽或十胜肽 以具有特別強的細胞毒殺性T淋巴球誘發能力。 201200525 本發明胜肽考慮經修飾的胜肽,其具有序列辨識號:2 至4之一胺基酸序列,其中一、二或多個胺基酸被取代或 加入’,、要經修飾之胜肽維持最初之細胞毒殺性T淋巴球 誘發能力。 此外’本發明提供編碼出任何本發明胜肽之經分離的 多核苷酸。這些多核苷酸可用以誘導具有細胞毒殺性τ淋 巴球誘發能力之抗原呈現細胞,或如本發明胜肽可被投予 至一個體以誘導抗癌症之免疫反應。 當投予至一個體時,本發明胜肽被表現於抗原呈現細 胞之表面以便誘導將分別之胜肽做為目標之細胞毒殺性τ 淋巴球。因此,本發明一目標為提供包括本發明任何之胜 肽或多核苷酸的試劑用以誘導細胞毒殺性Τ淋巴球。此 外,包括本發明任何之胜肽或多核苷酸的試劑、物質或組 合物可被用於癌症之治療及/或預防,及/或其手術後復發 的避免,此類癌症包括,但不限於睪丸腫瘤 '胰臟癌、膀 胱癌、非小細胞肺癌、小細胞肺癌與食道癌。因此,本發 明又另一目標為提供用於癌症之治療及/或預防,及/或其 手術後復發的避免的藥學試劑,此類試劑包括本發明任何 的胜肽或多核苷酸。代替本發明胜肽或多核苷酸/除了本發 明胜肽或多核苷酸外,本發明試劑或藥學試劑可包括呈現 任何之本發明胜肽的抗原呈現細胞或外吐小體為活性成 分。 本發明之胜肽或多核苷酸可被用來誘導於表面呈現 HLA抗原與一本發明胜肽之一複合物的抗原呈現細胞,例 201200525 如’藉由將來自一個體之抗原呈現細胞與此胜肽接觸或將 編碼出本發明胜肽的一多核苷酸引入抗原呈現細胞。此種 抗原呈現細胞具有高的抗目標胜肽之細胞毒殺性了淋巴球 誘發能力且因此對於癌症免疫治療為有用的。因此,本發 明另一目標為提供誘導具細胞毒殺性τ淋巴球誘發能力之 抗原呈現細胞的方法與藉由此方法獲得之抗原呈現細胞。 本發明更進一步之目標為提供誘導細胞毒殺性τ淋巴 球的方法,方法包括將CD8陽性細胞與表現一本發明胜肽 於其表面之抗原呈現細胞或外吐小體共培養之步驟,或引 入包括編碼出與本發明胜肽結合之了細胞受體(t cei上 receptor,TCR)次單元之多核苷酸的基因的步驟。藉由此 種方法可獲得之細胞毒殺性τ淋巴球也在癌症之治療及/ 或避免中提供用途,癌症之例子包括,但不限於睪丸腫瘤、 騰臟癌、膀胱癌' 非小細胞肺癌、小細胞肺癌與食道癌。 本發月另目標為提供藉由本發明方法可獲得之細 胞毒殺性Τ淋巴球。 本發明又另-目標提供於一需要之個體中誘導抗癌症 免疫反應的方法,此方法包括投予包含Μ肌2或其片 ^又1碼出MYBL2或其片段之多核苷酸,與呈現MYBL2或 、又之外吐小體或抗原呈現細胞之試劑或組合物的步 驟。 一:發明之應用性擴展至關於或起因於謂過度表現 ;、病的任個,包括癌症,其例子包括,但不限於 瘤姨臟癌、膀胱癌、非小細胞肺癌、小細胞肺癌 10 201200525 與食道癌。 更具體地,本發明提供下列事物: [1 ] 一種經分離的寡胜肽,包括一胺基酸序列,其係 擇自由序列辨識號:2至4所組成之群組,其中該寡胜肽 與一人類白血球組織抗原結合且具有細胞毒殺性T淋巴球 誘發能力。 [2] —種經分離的寡胜肽,其中該寡胜肽與一人類白 血球組織抗原結合且具有細胞毒殺性T淋巴球誘發能力, 其中該募胜肽係由擇自序列辨識號:2至4中之一胺基酸 序列所組成,其中1、2或數個胺基酸被插入、取代、刪除 或加入 [3] [2]之寡胜肽,其中該募胜肽具有下列特徵之一或 兩者: (a) 來自N端之第二個胺基酸係擇自白胺酸與曱硫丁 胺酸中;以及 (b) C端胺基酸係擇自纈氨酸與白胺酸中。 [4] [1]至[3]之經分離的寡胜肽’其中該人類白血球 組織抗原為人類白血球組織抗原-A2 [5] [1]至[4]之經分離的寡胜肽’其中該募胜肽為九 胜肽或十胜肽。 [6] —種經分離之多核苷酸,其編碼出[丨]至[5]之任 一項的寡胜肽。 [7 ] —種誘發細胞毒殺性τ淋巴球之試劑,其中該試 劑包括在[1 ]至[5 ]中之任一項所述之—或多個該募胜肽, 11 201200525 或在[6]中所述之一或多個該多核苷酸。 [8 ] —種藥學試劑,用於癌症之治療及/或預防,及/ 或其手術後復發的避免,其中該試劑包括在[1]至[5]中之 任一項所述之一或多個該寡胜肽,或在[6]中所述之一或多 個該多核苷酸。 [9 ][ 8 ]之藥學試劑,被配製來用以投予一個體,其人 類白血球組織抗原為人類白血球組織抗原-A 2。 [1 0 ][ 8 ]或[9 ]之藥學試劑,其被配製來用於治療癌 症。 [11 ] 一種誘導具有細胞毒殺性T淋巴球誘發能力之 抗原呈現細胞的方法,其中該方法包括下列步驟之一: (a) jy? hiro、ez 叩或/v? Wfo將一抗原呈現細 胞與[1]至[5]之任一項之寡胜肽接觸;或 (b) 將編碼出[1]至[5]之任一項之寡胜肽的一多核苦 酸引入一抗原呈現細胞。 [12 ] —種誘導細胞毒殺性T淋巴球的方法,藉由任何 之包括下列步驟之至少一個的方法: (a) 將CD8陽性T細胞與抗原呈現細胞共培養,抗原 呈現細胞表現一人類白血球組織抗原與[1 ]至[5 ]之任一項 之寡·胜狀的—複合物於其表面上; (b) 將CD8陽性τ細胞與外吐小體共培養,外吐小體 人 類白血球組織抗原與[1 ]至[5 ]之任一項之募胜肽 複合物於其表面上;以及 (c) 將一包括編碼出一 T細胞受體次單元多胜肽之多 12 201200525 w 核苦酸的基因引入一 T細胞,該T細胞受體次單元多胜肽 與[1]至[5]之任一項的寡胜肽結合。 [13 ]—種經分離之抗原呈現細胞,其表現一人類白血 球組織抗原與[1 ]至[5 ]之任一項之募胜肽的一複合物於其 表面上。 [1 4 ][ 1 3 ]之抗原呈現細胞,其藉由[11 ]之方法來誘 導。 [1 5 ]—種經分離之細胞毒殺性τ淋巴球,其以[1 ]至 [5]之任一項之胜肽為標的。 [1 6 ][ 1 5 ]之細胞毒殺性τ淋巴球,其藉由[1 2 ]之方法 來誘導。 [1 7] —種於一個體中誘導抗癌症之免疫反應的方 法’其包括下列步驟:投予該個體一試劑,該試劑包括[i ] 至[5 ]之任一項之寡胜肽、_其免疫活性片段,或一編碼出 該寡胜肽或免疫活性片段之一多核苷酸。 需瞭解的是,本發明前述發明概要與下列詳細敘述兩 者為示範之實施例,並不限制本發明或本發明其他替代實 施例。 除了上述,當以下詳細說明被閱讀並結合伴隨之圖式 與實施例,本發明之其他目的與特徵會變的更完全地明 白。然而,可瞭解的疋,上面之本發明内容與以下之詳細 說明兩者為示範之實施例,並不限制本發明或本發明其他 替代實施例。特別是,當關於-些特定實施例於此敛述之 本發明,可以瞭解的是,敘述為本發明之說明,且並不建 13 201200525 構為本發明之限制。各種修飾與應用可被熟悉此技藝人士 想到’而無背離本發明精神與範圍,如所时請專利範圍 所述。同樣地,本發明之其他目的 '特 付做、好處與優點自 此内容與下述之特定實施例,為清 M J芍/貧楚的,且對於熟悉此技 藝人士而言可立即明白。此種目#、特徵、好處與優點自 上述結合伴隨實施例、資料'圖式與所有要被自其單獨或 隨著考慮引人於此之參考文獻而描述的所有合理推論為清 楚的。 雖然於本發明實施例之實施或測試中可使用相似或等 同於在此敘述之那些的任何方法與材料,但是現在敘述較 佳之方法、元件與材料 '然而在敘述本發明材料與方法之 前’需瞭解的A ’本發明並不限於敘述於此之特定大小、 元狀尺寸#料、方法學、步驟等,例如按照慣例實驗 法及/或最佳化可將其變更。也需瞭解的是,於此敘述Μ 用之專門用語僅是為了敘述特別之變化形式或實施例,且 不傾向限制僅會受限於所附上之申請專利範圍的本 〜分刊物、專利或專利申請的 於此以其内容被具體引入A i V兩參考文獻。然而,於此並 被解釋為承S忍本發明由於务於政1丄 印A无則發明之效力不被給予先 些揭露之權力。 如果發生抵觸,本發日日Μ +赞明說明書,包括定義,將會控 14 201200525 此外,材料、方法與實施例僅為說明性,不意指被限制。 I.定義 於此使用之單字“一”與“該,,意指“至少—,,除非 以別的方式明確指出。 於此可替換使用之用語“多胜肽,,、“胜肽,,與“蛋 白質”意指胺基酸殘基之一聚合物。此用語適用於胺基酸 聚合物,於其中一或多個胺基酸殘基為經修飾之殘基或非 自然發生之殘基’例如對應自然發生胺基酸之人工化學模 仿物’與自然發生胺基酸聚合物。 於此使用之用語“胺基酸”意指自然發生與合成之胺 基酸’及胺基酸類似物與胺基酸模仿物,其與自然發生之 胺基酸起相似作用。自然發生胺基酸為基因密碼所編碼的 那些與於細胞中在轉譯後被修飾的那些(例如羥脯胺酸 (hydroxyproline) 、 r _ 羧基谷胺酸(gamma-08]^30乂7忍1111311^16)與〇-磷絲胺酸(〇11108?乜0361'1116))。 措辭“胺基酸類似物”意指具有與自然發生胺基酸相同之 基礎化學結構(一 &lt;2礙鍵結至一氫、一缓基、一胺基與一 R基)的化合物,但具有一經修飾之R基或經修飾之骨架 (例如,同絲胺酸(homoserine)、降亮胺酸(norleucine)、 甲硫胺酸(methionine)、亞颯(sulfoxide)、甲基硫氨續 (methionine methyl sul f on ium))。措辭“胺基酸模仿物” 意指化學化合物其與一般胺基酸具有不同結構,但有相似 的功能。 可藉由由 IUPAC-IUB Biochemical Nomenclature 15 201200525 C_issiGn所建議之其—般所知的三字母符號或-字母符 號來指出於此處之胺基酸。 ,於此可替換使用用語“基因”、“多核普酸”、“核Pilkinton U et a 1. , J Biol Chem 20 07 Jan 5 ; 282(1 ): 168-75). Furthermore, by analyzing the gene expression profile of a genome-wide DNA microarray containing 23,040 genes, it has been identified that MYBL2 is included in many cancers, including, for example, testicular tumors, pancreatic cancer, bladder cancer, non-small A novel molecule in which cell lung cancer, small cell lung cancer, and esophageal cancer are regulated upward (PTL 1, US 60/937, 61 6, PTL 2, WO 2004/031410). MYBL2 is considered to be a novel oncoant antigen (oncoant igen), and the epitope peptide derived from MYBL2 can be applied to cancer immunotherapy in patients with several forms of tumor. [Citations] Patent Literature [PTL 1] US 60/937, 616 [PTL 2] WO 2004/031410 201200525 Non Patent Literature [NPL 1] Boon T, Int J Cancer 1993 May 8 , 54(2): 177-80 [NPL 2] Boon T &amp; van der Bruggen P, J Exp Med 1996 Mar 1, 183(3): 725-9 [NPL 3] Harris CC, J Natl Cancer Inst 1 996 Oct 16, 88(20): 1442-55 [NPL 4] Butterfield LH et al. , Cancer Res 1 999 Jul 1, 59(13): 3134-42 [NPL 5] Vissers JL et al., Cancer Res 1999 Nov 1, 59(21): 5554-9 [NPL 6] van der Burg SH et al. , J Immunol 1 996 May 1, 156(9): 3308-14 [NPL 7] Tanaka F et al., Cancer Res 1 997 Oct 15, 57(20): 4465-8 [NPL 8] Fujie T et al., Int J Cancer 1 999 Jan 18, 80(2): 169-72 [NPL 9] Kikuchi M et al., Int J Cancer 1999 May 5, 81(3): 459-66 [NPL 10] Oiso M et al., Int J Cancer 1 999 May 5, 81(3): 387-94 [NPL 11] Belli F et al., J Clin Oncol 2002 Oct 15, 20(20): 4169-80 [NPL 12] Coulie PG et al., Immunol Rev 2002 Oct, 5 201200525 188: 33-42 [NPL 13] Rosenberg SA et al., Nat Med 20 04 Sep, 10(9): 909-15 [NPL 14] Nomura N et al., Nucleic Acids Res. 1988 Dec 9, 1 6(23): 1 1 075 - 1 1 089 [NPL 15] Robinson C et al Oncogene 1 996 May 2; 12(9):1855-64 [NPL 16] Lane et al. , Oncogene 1997 May 22; 14(20): 2445-53 [NPL 17] Sala et al., Proc Natl Acad Sci 1 997 Jan 21; 94(2): 532 - 536 [NPL 18] Johnson K et al., J Biol Chem 1999 Dec 17;274(51):36741-9 [NPL 19] Pilkinton M et al., J Biol Chem 2007 Jan 5:282(1): 168-75 SUMMARY OF THE INVENTION The present invention is based, in part, on the discovery of suitable targets for immunotherapy. Since tumor-associated antigens (TAAs) are sometimes perceived by the immune system as "self" and therefore often have no immunogenicity, the discovery of suitable targets is extremely important. As mentioned above, it has been confirmed that MYBL2 (sequence number encoded by GenBank numbered _002466 (SEQ ID NO: 5) is encoded by the gene: 6) in cancers such as sputum tumor, pancreatic cancer, bladder cancer Small cell lung cancer, 201200525 Small cell lung cancer and esophageal cancer tissue are up-regulated. Therefore, MYBL2 is a candidate for cancer/tumor immunotherapy. The present invention is based, at least in part, on the recognition of specific epitope peptides of MYBL2 which have the ability to induce cytotoxic T lymphocytes specific for MYBL2. As discussed in detail below, a candidate peptide from MYBL2 that binds to Hu (human leukocyte antigen)-A2 is used to stimulate peripheral blood mononuclear cells (peripheral M〇C) d mc)nc) from a health provider. ]_ cel Is, PBMCs). Specifically, a cytotoxic cytotoxic T lymphocyte having a specific cytotoxicity against HLA-A2 positive target cells pulsed by each candidate peptide was established. Thus, these results demonstrate that these traits are HLA-A2-restricted epitopes, which induce a strong and specific immune response against cells expressing MYBL2. Therefore, these results show that Μγβ [2 is potently immunogenic and its epitope is a valid target for tumor immunotherapy. Accordingly, it is an object of the present invention to provide an isolated peptide which binds to an HLA antigen, which is MYBL2 (SEQ ID NO: 6) or a fragment thereof. These traits are expected to have cytotoxic T lymphocyte evokengs and can therefore be used to induce cytotoxic thymocytes or can be administered to a body to induce an immune response against cancer, cancer Examples include, but are not limited to, testicular tumors, smear cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. Preferably, the peptides are a nine-peptide or a ten-peptide, and more preferably have an amino acid sequence selected from the sequence identification number: 2 to 4, particularly the sequence identification number: 3 The nine peptide or the ten peptide of the amino acid sequence has a particularly strong cytotoxic T lymphocyte evoking ability. 201200525 The peptide of the present invention considers a modified peptide having a sequence identification number of 2 to 4 amino acid sequence in which one, two or more amino acids are substituted or added, and the modification is to be successful. The peptide maintains the original cellular toxic T lymphocyte evoking ability. Further, the invention provides isolated polynucleotides encoding any of the peptides of the invention. These polynucleotides can be used to induce antigen-presenting cells having cytotoxic cadmium-inducing ability, or as the peptide of the present invention can be administered to a body to induce an immune response against cancer. When administered to a body, the peptide of the present invention is expressed on the surface of the antigen-presenting cell to induce a cytotoxic tau lymphocyte to which the respective peptide is targeted. Accordingly, it is an object of the present invention to provide an agent comprising any of the peptides or polynucleotides of the present invention for inducing cytotoxic lymphocytes. Furthermore, an agent, substance or composition comprising any of the peptides or polynucleotides of the invention can be used for the treatment and/or prevention of cancer, and/or its avoidance of recurrence after surgery, including but not limited to such cancers Sputum tumors, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer and esophageal cancer. Accordingly, it is yet another object of the present invention to provide pharmaceutical agents for the treatment and/or prevention of cancer, and/or the avoidance of recurrence after surgery, such agents comprising any of the peptides or polynucleotides of the present invention. In place of the peptide or polynucleotide of the present invention/except for the peptide or polynucleotide of the present invention, the agent or pharmaceutical agent of the present invention may comprise an antigen-presenting cell or an exosome which exhibits any of the peptides of the present invention as an active ingredient. The peptide or polynucleotide of the present invention can be used to induce an antigen-presenting cell which exhibits a complex of a HLA antigen and a peptide of the present invention on the surface, for example, 201200525, such as by presenting an antigen from a body to a cell. The peptide is contacted or a polynucleotide encoding the peptide of the present invention is introduced into the antigen-presenting cell. Such antigen-presenting cells have a high anti-target peptide cell toxic lymphocytosis-inducing ability and are therefore useful for cancer immunotherapy. Therefore, another object of the present invention is to provide a method for inducing an antigen-presenting cell having a cytotoxic lymphoblast-inducing ability and an antigen-presenting cell obtained by the method. A still further object of the present invention is to provide a method for inducing a cytotoxic tau lymphocyte, the method comprising the steps of co-cultivating a CD8-positive cell with an antigen-presenting cell or an exosome expressing a peptide of the invention, or introducing A step of encoding a gene encoding a polynucleotide of a cellular receptor (t cei receptor, TCR) subunit in combination with a peptide of the present invention. The cytotoxic tau lymphocytes obtainable by such methods are also useful in the treatment and/or avoidance of cancer, and examples of cancer include, but are not limited to, testicular tumors, smear cancer, bladder cancer 'non-small cell lung cancer, Small cell lung cancer and esophageal cancer. Another goal of this month is to provide a cytotoxic sputum lymphocyte obtainable by the method of the present invention. Still another object of the present invention is to provide a method for inducing an anti-cancer immune response in a subject in need thereof, which comprises administering a polynucleotide comprising a diaphragm 2 or a fragment thereof and a code of MYBL2 or a fragment thereof, and presenting MYBL2 Or the step of exosome or antigen presenting a reagent or composition of the cell. One: the applicability of the invention extends to or is caused by excessive performance; any of the diseases, including cancer, examples including, but not limited to, tumor sputum cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer 10 201200525 With esophageal cancer. More specifically, the present invention provides the following: [1] An isolated oligopeptide comprising an amino acid sequence selected from the group consisting of 2 to 4, wherein the oligopeptide It binds to a human leukocyte tissue antigen and has a cytotoxic T lymphocyte evoking ability. [2] an isolated oligopeptide, wherein the oligopeptide binds to a human leukocyte antigen and has cytotoxic T lymphocyte evoking ability, wherein the peptide is selected from sequence identification number: 2 to One of amino acid sequences of 4, wherein 1, 2 or several amino acids are inserted, substituted, deleted or added to the oligopeptide of [3] [2], wherein the recruit peptide has one of the following characteristics Or both: (a) the second amino acid from the N-terminus is selected from the group consisting of leucine and acesulfame; and (b) the C-terminal amino acid is selected from the group consisting of proline and leucine . [4] The isolated oligopeptide of [1] to [3] wherein the human leukocyte antigen is an isolated oligopeptide of human leukocyte antigen-A2 [5] [1] to [4] The recruit peptide is a nine-peptide or a ten-peptide. [6] An isolated polynucleotide encoding the oligopeptide of any one of [丨] to [5]. [7] An agent for inducing a cytotoxic thymocyte, wherein the agent comprises any one of [1] to [5] - or a plurality of the peptides, 11 201200525 or at [6] One or more of the polynucleotides described in the . [8] a pharmaceutical agent for the treatment and/or prevention of cancer, and/or prevention of recurrence after surgery, wherein the agent comprises one of any one of [1] to [5] or a plurality of the oligopeptides, or one or more of the polynucleotides described in [6]. The pharmaceutical agent of [9] [8] is formulated for administration to a human, and the human leukocyte tissue antigen is human leukocyte antigen-A 2 . A pharmaceutical agent of [1 0 ] [8] or [9] which is formulated for the treatment of cancer. [11] A method of inducing an antigen-presenting cell having a cytotoxic T lymphocyte-inducing ability, wherein the method comprises one of the following steps: (a) jy? hiro, ez 叩 or /v? Wfo presents an antigen to cells and The oligopeptide contact of any one of [1] to [5]; or (b) introducing a polynucleic acid encoding the oligopeptide of any one of [1] to [5] into an antigen-presenting cell . [12] A method for inducing cytotoxic T lymphocytes by any method comprising at least one of the following steps: (a) co-culturing CD8-positive T cells with antigen-presenting cells, the antigen-presenting cells exhibiting a human leukocyte The tissue antigen and the oligo-synthesis of any one of [1] to [5] are on the surface thereof; (b) the CD8-positive tau cells are co-cultured with the exosome, and the human white blood cells are exosome a tissue antigen and a peptide complex of any one of [1] to [5] on its surface; and (c) a nucleic acid comprising a T cell receptor subunit multi-peptide 12 201200525 w nucleus The bitter acid gene is introduced into a T cell which binds to the oligopeptide of any one of [1] to [5]. [13] An isolated antigen-presenting cell which exhibits a complex of a human leukocyte antigen and a peptide of any one of [1] to [5] on its surface. The antigen of [1 4 ][ 1 3 ] presents cells which are induced by the method of [11]. [1 5 ] An isolated cytotoxic tau lymphocyte, which is characterized by a peptide of any one of [1] to [5]. [1 6 ] [ 1 5 ] cytotoxic τ lymphocytes, which are induced by the method of [1 2 ]. [1 7] A method for inducing an immune response against cancer in a body, which comprises the step of administering to the individual a reagent comprising the oligopeptide of any one of [i] to [5], An immunologically active fragment thereof, or a polynucleotide encoding one of the oligopeptide or immunologically active fragment. It is to be understood that the invention is not intended to In addition to the above, other objects and features of the present invention will become more fully apparent from the following detailed description. However, the present invention and the following detailed description are exemplary embodiments of the invention, and are not intended to limit the invention or other alternative embodiments of the invention. In particular, the invention as set forth in the Detailed Description of the Invention is to be understood as illustrative of the invention and is not to be construed as a limitation. Various modifications and applications can be made by those skilled in the art without departing from the spirit and scope of the invention, as described in the scope of the claims. Likewise, other objects, advantages, advantages and advantages of the invention are apparent from the following description and the particular embodiments set forth below, and are readily understood by those skilled in the art. Such terms, features, advantages and advantages are apparent from the above-described combination of the accompanying examples, the <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> and all reasonable inferences that are to be described as a separate reference. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the embodiments of the present invention, the preferred methods, elements and materials are now described, however, prior to describing the materials and methods of the present invention, The invention is not limited to the particular size, dimensions, methods, steps, etc. described herein, such as may be modified by conventional experimentation and/or optimization. It is also to be understood that the specific terms used herein are for the purpose of describing particular variations or embodiments and are not intended to limit the scope of the invention, The patent application is hereby specifically incorporated into the A i V two references. However, this is also interpreted as the intent of the invention. The effect of the invention is not given the prior disclosure. In the event of a conflict, the date of this issue + praise of the instructions, including definitions, will be controlled 14 201200525 In addition, the materials, methods and examples are illustrative only and are not intended to be limiting. I. Definitions The words "a" and "the", as used herein, mean "at least-, unless explicitly stated otherwise. The phrase "polypeptide," "peptide," and "protein" means a polymer of one of the amino acid residues. This term applies to amino acid polymers in which one or more amino acid residues are modified residues or non-naturally occurring residues 'for example, artificial chemical mimics corresponding to naturally occurring amino acids' and nature An amino acid polymer is produced. As used herein, the term "amino acid" means naturally occurring and synthetic amino acid' and an amino acid analog and an amino acid mimetic which function similarly to naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code and those modified in the cell after translation (eg hydroxyproline, r _ carboxy glutamate (gamma-08) ^ 30 乂 7 tolerate 1111311 ^16) with 〇-phosphoric acid (〇11108?乜0361'1116)). The expression "amino acid analog" means a compound having the same basic chemical structure as the naturally occurring amino acid (a &lt; 2 barrier to a hydrogen, a slow group, an amine group and an R group), but Having a modified R group or modified backbone (eg, homoserine, norleucine, methionine, sulfoxide, methyl sulfoxide) Methionine methyl sul f on ium)). The expression "amino acid mimetic" means a chemical compound which has a different structure from a general amino acid but which has a similar function. The amino acid herein can be indicated by the three-letter symbol or the letter symbol generally known by IUPAC-IUB Biochemical Nomenclature 15 201200525 C_issiGn. Here, the terms "gene", "multi-nucleotide", "nuclear" may be used interchangeably.

苷酸與“核酸”,且除非U 、 別的特別方式指出,相似於 胺基酸其以它們-般被接受的單―字母編碼來指[ 此處可替換使用之用語“試劑,’肖“組合物,,意指一 產物,其包括於特定量中之特定成分,與任何產物其直接 或間接來自於特定量之特定士八 、疋成刀的組合。此用語當使用於 與“藥學試劑”與“藥學組合物,,相關,意指包括一產 物’其包括-活性成分與任何形成載體的惰性成分,及任 何產物其直接或間接來自任兩個或多個成份之組合、複人 或聚集,或來自一或多個成分之解離,或來自-或多個成 分之反應或相互作用的其他形式,此,在本發明内容中 用語“藥學試劑,,肖“藥學組合物,,意指藉Μ合本’ 分子或化合物與藥學上或生理上可接受之載體所 何產物。如此處所使用之措辭“藥學上可接受 ,, “生理上可接受之載體”意指藥學上或生理上…或 料、組合物、物質或載劑,包括,但不限於-液體=之材 填充料、稀釋劑、賦形劑、溶劑或套膜材料,=固體 官或身體之一部分攜帶或運輸受支配之支架、益 (scaffolded p〇lypharmac〇ph〇res)至另一 ^ 效團 一部分相關。 s身體之 於此使用之用語“活性成分,,意指在-試劑或叫 物中的物質,其為生物或生理活躍的。特3 ,= ^ ° 疋,在一藥學 16 201200525 試劑或組合物中“活性成分”意指一物質其顯示一目標的 之藥學作用。例如’若藥學試劑或組合物用於癌症之治療 或避免中’在試劑或組合物中的活性成分可直接或間接引 起對癌細胞及/或組織的生物或生理作用。較佳為,此作用 可包括減低或抑制癌細胞生長、損傷或殺死癌細胞及/或組 織等。一般而言,有效成分的間接作用為誘導細胞毒殺性 τ細胞辨認或殺死癌細胞。在配製前,“活性成分”也意 指為“主體(bulk)” 、 “藥物物質(drugsubstance)”戒 技術產物(technical product),,。 除非以別的方式定義,用語“癌症,,意指過度表現 MYBL2基因之癌症,其例子包括,但不限於睪丸腫瘤、胰 臟癌、膀胱癌、非小細胞肺癌、小細胞肺癌與食道癌。 除非以別的方式定義,於此可替換使用且除非以別的 方式特別指出之用語“細胞毒殺性τ淋巴球,,、“細胞毒 殺性τ細胞,,與“CTLV以意指了淋巴球之次族群,意指了 淋巴球之次群組(sub_group) ’其可辨認非自身細胞(例 如’腫瘤細胞、被病毒感染之細胞),且誘導這些細胞死 亡。 二非特別定義,用語“HLA_A〇2 ”意、指包含次型,例如 1201 或 HLA- A*-0206 之 HLA-A2。 “治=於本發明之方法與組合物之範圍提供用途於癌症之 臨床優:内容而言’一治療被視為“有效’,,若其導致 中癌症 錢中的減少、或於個體 小、普遍程度(Prevalence)或轉移潛力的滅 17 201200525 少。當治療為預防性(pr0phy iact ical ly)提供時,“有效” 意指減緩或避免癌症形成,或避免或減輕癌症之臨床症 狀。有效性被確認於相關之診斷或治療特定腫瘤形式的任 何已知方法β 除非特別定義,於此使用之用語“套組”被使用於關 於試劑與其他材料之組合。與此考慮之套組包括微陣列、 阳片、標誌等。並無打算使用語“套組”限制於試劑及/ 或材料之特定組合。 如於此使用,在一個體或病患的背景中,措辭‘‘ 陽性”意指此個體或病患同型結合地(h〇m〇zyg〇usly)或異 質結合地(heter0zyg0USly)具有HLA_A2抗原基因,且Glycosidic acid and "nucleic acid", and unless otherwise indicated by U, other special methods, similar to amino acids, which are referred to by their commonly accepted mono-letter codes. [herein the term "replaceable" reagent, '肖' By composition, it is meant a product which is included in a particular amount of a particular ingredient, in combination with any product which is derived directly or indirectly from a particular amount of a particular octant. This terminology, when used in connection with "pharmaceutical agent" and "pharmaceutical composition, is meant to include a product which includes - the active ingredient with any inert ingredient which forms a carrier, and any product which is derived directly or indirectly from either or both Combinations of multiple components, complex or aggregated, or dissociation from one or more components, or other forms of reaction or interaction from - or multiple components, as used herein, the term "pharmaceutical reagents," "Pharmaceutical composition," means a product of a molecule or compound and a pharmaceutically or physiologically acceptable carrier. As used herein, the phrase "pharmaceutically acceptable," physiologically acceptable carrier "meaning pharmaceutically or physiologically... or a material, composition, substance or carrier, including, but not limited to, - liquid = material filler, diluent, excipient, solvent or sheath material, = solid official or One part of the body carries or transports a controlled scaffold, scaffolded p〇lypharmac〇ph〇res to another part of the effect. The term "active ingredient" as used herein refers to a substance in a reagent or a substance that is biologically or physiologically active. Special 3, = ^ ° 疋, in a pharmacy 16 201200525 Reagent or composition By "active ingredient" is meant a pharmaceutical substance which exhibits a pharmaceutical effect. For example, if a pharmaceutical agent or composition is used in the treatment or avoidance of cancer, the active ingredient in the agent or composition may directly or indirectly cause Biological or physiological action of cancer cells and/or tissues. Preferably, the action may include reducing or inhibiting the growth of cancer cells, damaging or killing cancer cells and/or tissues, etc. In general, the indirect action of the active ingredients is induction. Cytotoxic tau cells recognize or kill cancer cells. Before preparation, "active ingredient" also means "bulk", "drug substance" or technical product, unless Other definitions, the term "cancer," means cancer that overexpresses the MYBL2 gene, examples of which include, but are not limited to, testicular tumors, pancreatic cancer, bladder cancer, non-small cell lungs , Small cell lung cancer and esophageal cancer. Unless otherwise defined, it may be used interchangeably and unless otherwise specifically indicated by the term "cytotoxic tau lymphocytes," "cytotoxic tau cells," and "CTLV to mean lymphocytes. A subgroup refers to a subgroup of lymphocytes (sub_group) that recognizes non-self cells (eg, 'tumor cells, cells infected with the virus) and induces death of these cells. 2. Not specifically defined, the term "HLA_A〇 2 ” means “HLA-A2 containing a subtype, such as 1201 or HLA-A*-0206. “Treatment = provides a clinical use of cancer in the context of the methods and compositions of the present invention: content It is considered “effective” if it causes a reduction in cancer money, or a small, prevalence or metastatic potential of the individual. 201200525 Less when the treatment is provided for preventive (pr0phy itical ly) "Effective" means slowing or avoiding the formation of cancer, or avoiding or ameliorating the clinical symptoms of cancer. Validity is identified in any known method of diagnosis or treatment of a specific form of tumor beta unless special The term "set" as used herein is used in connection with reagents and other materials. The set considered here includes microarrays, positives, markers, etc. It is not intended to use the term "set" to limit reagents. And/or a specific combination of materials. As used herein, in the context of a subject or patient, the word ''positive') means that the individual or patient has the same type of binding (h〇m〇zyg〇usly) or heterogeneous binding. (heter0zyg0USly) has the HLA_A2 antigen gene, and

HlA_A2抗原被表現於此個體或病患的細胞中為一 hla抗 原。 “對於本發明之物質與方法之範圍提供用途於癌症之 避免與預防之内容而言,此類用詞為與此交替使 用意指任何活性,其減少死亡率之貞載或來自疾病之死亡 率。避免與預防可發生力“初期、第二期與第三期避免層 級’,。初期避免與預防避免了疾病之發展,而第二期與第 三期層級之避免與預防包括藉由恢復功能與減少疾病相關 併發症’以疾病之發展與症狀漳 対低/、征狀疋年現及減少已建立之疾病 之負向發展的避免盘預ρ方為,, '、頂防馬目的。或者,治療或避免可包 括一廣範圍之預防疾病治療,其 陳共以減緩特別疾病之嚴重度 為目標,例如減少腫瘤之增殖與轉移。 在本發明内容中,癌症之、、二,佐η / 烛屁之/α療及/或預防,及/或其手 18 201200525 術後復發的避免包括任何下列步驟,例如癌細胞之手術移 除:似癌細胞之生長抑制、腫瘤之衰退或退化、癌發生之 減緩與抑制的誘導,與轉移之減少與抑制。癌症之有效治 療及/或預防減少致死率與改善具有癌症之個體的預後、減 低癌症標記於血液中的程度與減緩伴隨著癌症之可偵測症 狀。例如,症狀之減輕或改善構成有效治療及/或預防,其 包括10%、20%、30%或更加減輕,或穩定疾病。 除非特別定義,於此使用之所有技術或科學用語為與 屬與本發明之熟悉此技藝人士所通常瞭解之意義相同。 11.胜肽 為了也明來自MYBL2之胜狀作用如一被細胞毒殺性τ 淋巴球(CTLs)所辨認之抗原,分析來自MYBL2之胜肽(序 列辨識號:6 )以確定是否其為由一般遇到HLA對偶基因 (a 11 e 1 e)之HLA (人類白血球組織抗原)-a2所限制之抗 原決定位(Date Y ei a/.,Tissue Antigens 47: 93-101, 1996; Kondo A et al., J Immunol 155: 4307-12, 1995; Kubo RT a/., J Immunol 152: 391 3-24,1 994)。確認 來自MYBL2之HLA-A2結合胜肽的候選物,根據其對HLA-A2 之結合親和力。下列胜肽被認為是候選胜肽: MYBL2-A02-9-597 (序列辨識號:2)、 MYBL2-A02-9-1 44 (序列辨識號:3)與 MYBL2-A02-10-596 C 序列辨識號:4)。 i/7 h 藉由載有這些胜肽之樹突細胞(dendr i t i c cel 1,DC)刺激T細胞後,使用下列胜肽成功建立細胞毒殺 19 201200525 性τ淋巴球: MYBL2-A02-9-144 (序列辨識號:3) 這些被建立的細胞毒殺性τ淋巴球顯示強而專一之抗 經分別之胜肽脈衝之目標細胞的細胞毒殺性τ淋巴球活 性。此處這些結果證明MYBL2為一由細胞毒殺性Τ淋巴球 所辨認之抗原,且胜肽為由HLA-A2限制之MYBL2的抗原決 定位胜肽。 由於MYBL2基因於癌症細胞與組織,包括,但不限於 例如睪丸腫瘤、胰臟癌、膀胱癌、非小細胞肺癌、小細胞 肺癌與食道癌的那些之中被過度表現,且不被表現於大部 分正常器官中,所以其構成良好之免疫治療標的。因此, 本發明提供對應於MYBL2之細胞毒殺性Τ淋巴球辨認的抗 原決定位的九胜肽(胜肽由九個胺基酸殘基所組成)與十 胜肽(胜肽由十個胺基酸殘基所組成)^本發明九胜肽與 十胜肽之較佳實施例包括具有擇自於序列辨識號:2至4 中’特別是3之胺基酸序列的那些胜肽。The HlA_A2 antigen is expressed in the cells of the individual or patient as a hla antigen. "For the context of the use of the substances and methods of the invention to provide for the avoidance and prevention of cancer, such terms are used interchangeably to mean any activity which reduces mortality or mortality from disease. Avoidance and prevention can occur in the "initial, second and third phase avoiding hierarchy". Early avoidance and prevention avoids the development of the disease, while the avoidance and prevention of the second and third stages includes the restoration of function and the reduction of disease-related complications, and the development and symptoms of the disease are degraded/symptoms. Now, and to reduce the negative development of the established disease, the avoidance of the pre-requisites, ', top anti-horse. Alternatively, treatment or avoidance may include a wide range of disease prevention treatments, with the goal of slowing the severity of particular diseases, such as reducing tumor proliferation and metastasis. In the context of the present invention, the avoidance of postoperative recurrence of cancer, bis, argon/cafe/alpha therapy and/or prevention, and/or its hand 18 201200525 includes any of the following steps, such as surgical removal of cancer cells : growth inhibition of cancer cells, tumor regression or degeneration, induction of slowing and inhibition of cancer, and reduction and inhibition of metastasis. Effective treatment and/or prevention of cancer reduces mortality and improves the prognosis of individuals with cancer, reduces the extent to which cancer marks in the blood, and slows the detectable symptoms associated with cancer. For example, amelioration or amelioration of symptoms constitutes effective treatment and/or prevention, including 10%, 20%, 30% or less, or stabilization of the disease. All technical or scientific terms used herein have the same meaning as commonly understood by those skilled in the art, unless otherwise defined. 11. Peptide In order to also recognize the phenotype of MYBL2, such as an antigen recognized by cytotoxic T lymphocytes (CTLs), the peptide derived from MYBL2 (SEQ ID NO: 6) was analyzed to determine whether it was a general condition. To the HLA (a 11 e 1 e) HLA (human leukocyte antigen)-a2 restricted epitope (Date Y ei a /., Tissue Antigens 47: 93-101, 1996; Kondo A et al. , J Immunol 155: 4307-12, 1995; Kubo RT a/., J Immunol 152: 391 3-24, 1 994). Candidates for HLA-A2 binding peptides from MYBL2 were confirmed based on their binding affinity for HLA-A2. The following peptides are considered to be candidate peptides: MYBL2-A02-9-597 (SEQ ID NO: 2), MYBL2-A02-9-1 44 (SEQ ID NO: 3) and MYBL2-A02-10-596 C sequence Identification number: 4). i/7 h After stimulating T cells by dendritic cells (dendr itic cel 1, DC) carrying these peptides, the following peptides were used to successfully establish cytotoxicity 19 201200525 τ lymphocytes: MYBL2-A02-9-144 (SEQ ID NO: 3) These established cytotoxic τ lymphocytes showed a strong and specific cytotoxic thymocyte activity against the target cells of the respective peptides. Here, these results demonstrate that MYBL2 is an antigen recognized by the cytotoxic lymphocytes, and the peptide is an antigen-resolved peptide of MYBL2 restricted by HLA-A2. Since the MYBL2 gene is overexpressed in cancer cells and tissues, including but not limited to, for example, testicular tumors, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer, and is not expressed in large Part of the normal organs, so it constitutes a good immunotherapeutic target. Accordingly, the present invention provides a nine-peptide (the peptide consists of nine amino acid residues) and ten peptides (the peptide consists of ten amino groups) corresponding to the epitope of the cytotoxic lymphocyte recognition of MYBL2. Preferred Examples of the Nine Peptides and Decapeptides of the Invention include those peptides having the amino acid sequence of 'Special 3' selected in Sequence Numbers: 2 to 4.

通常可使用現今於例如網路可得之軟體程式,例如於 Parker KC et al. , J Immunol 1994 Jan 1, 152(1): 163-75 中所敘述的那些,來計算介於各種胜肽與hla 抗原間之結合親和力。例如,參照parker kc aA,JIt is common to use software programs now available, for example, on the Internet, such as those described in Parker KC et al., J Immunol 1994 Jan 1, 152(1): 163-75, to calculate between various peptides and Binding affinity between hla antigens. For example, refer to parker kc aA, J

Immunol 1994 Jan 1,1 52( 1 ): 163-75;與 Kuzushima K a/., Blood 2001, 98(6): 1872-81 中所述可測量與 HLA 抗原之結合親和力。測量親和力之方法敘述,例如於 Journal of Immunological Methods, 1995, 185: 181-190 20 201200525 與 Protein Science,2000,9: 1838-1 846 中。所以使用 此種軟體程式可選擇來自MYBL2的片段,其具有與Hu抗 原之高結合親和力。因此本發明包括由來自MYBL2之任何 片所組成之胜肽,其藉由使用此類已知程式確認與Η&quot; 結合。本發明胜肽可由全長之mYBL2所組成。 本發明胜肽’較佳為本發明九胜肽與十胜肽可於側面 具有額外之胺基酸殘基,只要所產生之胜肽維持它們的細 胞毋殺性T淋巴球誘發能力。位於本發明胜肽側面之胺基 酸殘基可由任何種類之胺基酸所組成,只它們不減少原始 胜肽之細胞毒殺性τ淋巴球誘發能力。因此,本發明包含 胜肽,其包括來自MYBL2之胜肽且具有對HLA抗原之結合 親和力。此種胜肽’一般小於約4 〇個胺基酸,時常小於約 20個胺基酸’通常小於約15個胺基酸。 一般而言,於一胜肽中一、二或多個胺基酸之修飾, 不會影響胜肽的功能,且在一些例子中,甚至增強原始蛋 白質所需之功能。事實上’已知經修飾之胜肽(即,胜肽 包括當與原始參考序列比較時於其中一、二或多個胺基酸 殘基已被修飾(即’取代、刪除、加入或插入)的胺基酸 序列)維持原始胜肽的生物活性(Mark et al.,Proc Nat 1 Acad Sci USA 1984, 81: 5662-6; Zoller and Smith,Immunol 1994 Jan 1, 1 52(1): 163-75; The binding affinity to HLA antigens can be measured as described in Kuzushima Ka/., Blood 2001, 98(6): 1872-81. A method for measuring affinity is described, for example, in Journal of Immunological Methods, 1995, 185: 181-190 20 201200525 and Protein Science, 2000, 9: 1838-1 846. Therefore, a fragment derived from MYBL2, which has a high binding affinity to Hu antigen, can be selected using this software program. The invention thus encompasses peptides consisting of any of the fragments from MYBL2, which are confirmed to be combined with Η&quot; by using such known programs. The peptide of the present invention can be composed of full length mYBL2. Preferably, the peptide P of the present invention has the additional amino acid residues on the side of the nine peptides and the ten peptides of the present invention as long as the peptides produced maintain their cell killing T lymphocyte inducing ability. The amino acid residues located on the side of the peptide of the present invention may be composed of any kind of amino acid, and they do not reduce the cytotoxic lytic lymphocyte inducing ability of the original peptide. Thus, the invention encompasses a peptide comprising a peptide derived from MYBL2 and having a binding affinity for an HLA antigen. Such peptides are generally less than about 4 amino acids, often less than about 20 amino acids, typically less than about 15 amino acids. In general, the modification of one, two or more amino acids in a peptide does not affect the function of the peptide and, in some instances, even enhances the desired function of the original protein. In fact 'known modified peptides (ie, peptides include one, two or more amino acid residues that have been modified (ie, substituted, deleted, added or inserted) when compared to the original reference sequence The amino acid sequence) maintains the biological activity of the original peptide (Mark et al., Proc Nat 1 Acad Sci USA 1984, 81: 5662-6; Zoller and Smith,

Nucleic Acids Res 1982, 10: 6487-500; Dalbadie-Nucleic Acids Res 1982, 10: 6487-500; Dalbadie-

McFarland et al., Proc Natl Acad Sci USA 1982, 79: 6409-13)。因此’於一實施例中,本發明胜肽具有細胞毒 殺性T淋巴球誘發能力與擇自序列辨識號:2至4中,特 21 201200525 別是3之胺基酸序列兩者,其中加入、插入、刪除及/或取 代一、二甚至更多個胺基酸。 熟悉此技藝人士認定改變一單一胺基酸或一小百分比 之胺基酸之個別加入或取代至一胺基酸序列傾向產生保存 原始胺基酸支鏈的特性。因此,它們常被意指為“保守取 代(conservative substitutions)” 或“保守修飾 (conservative modifications)” ,其中一蛋白質之改變 導致一具有類似原始蛋白質之功能的經修飾蛋白質。提供 功能相似胺基酸之保守取代表已為本技術領域所熟知。所 需保寸之胺基酸支鍵的特徵的例子包括,例如疏水胺基酸 (A,I,L,M,F,P,W,Y,V)、親水胺基酸(R,D, N, C, E,Q,G,H,K, S,T)與具有下列共同官能基或特徵之支 鏈:一脂肪族支鏈(G,A,V,L,I, P); —含羥基支鏈(S, Τ, Y);含硫原子支鏈(C,M);含羧酸與胺基支鏈(D,N,E, Q);含鹼支鏈(R,K,H);以及含芳香族支鏈(H,F, Y,W)。 此外’下列八個族群各包含於本技術領域中被接受為保守 取代之胺基酸: 1) 丙胺酸(A)、甘胺酸(G); 2) 天門冬胺酸(D)、麩胺酸(E); 3) 天門冬醯胺(N)、麩胺醯胺(Q); 4) 精胺酸(R)、離胺酸(K); 5) 異白胺酸(I)、白胺酸(L)、曱硫丁胺酸(M)、纈胺 酸(V); 6) 笨丙胺酸(F)、酪胺酸(Y)、色胺酸(w); ⑤ 201200525 7) 絲胺酸(S)、蘇胺酸(Τ);以及 8) 半胱胺酸(C)、甲硫丁胺酸(Μ)(參見,例如 Creighton, Proteins 1984)。 此種經保守修飾胜肽也被視為本發明之胜肽。然而, 本發明之胜肽並不限於此,且可包括非保守修飾,只要所 產生經修飾之胜肽維持原始胜肽之細胞毒殺性T淋巴球誘 發能力。更進一步而言,經修飾之胜肽不排除多形變體 (polymorphic variant)、種間同質體(interspecies homologues)與MYBL2對偶基因(alleles)之細胞毒殺性τ 淋巴球誘發的胜肽。 為了維持必須之細胞毒殺性T淋巴球誘發能力,可修 飾(插入 '刪除、加入及/或取代)一小數目(例如一、二 或數個)或小百分比之胺基酸。此處用語“數個,,指5或 更少個胺基酸,例如4個、3個或更少。要被修飾之胺基 酉文之百分比較佳為2〇%或更少’更佳為15%或更少’且甚 至更佳為,10%或更少,或1至5%。 此外,本發明胜肽可被以胺基酸殘基插入、取代或加 入或胺基酸殘基可被刪除以達到一較高之親和力。當使用 於文中之免疫治療時,本發明之胜肽應被表現於一細胞或 外吐小體之表面上,較佳作為一具有Hu抗原之複合物。 因此較佳為選擇胜肽其不止誘導細胞毒殺性τ淋巴球也 具有對HLA抗原之高親和力。為達此目的,㈣可藉由胺 基酸殘基之取代、插人及/或加入來修飾以產生具經改善之 結合親和力的經修饰之胜肽。除了自然表現之胜肽外,由 23 201200525 於已知藉由結合至HLA抗原表現之胜肽序列的規則(j Immunol 1994, 152: 3913; Immunogenetics 1995, 41: 178; J Immunol 1994,155 : 4307) ’因此可將基於此規則之修 飾引入本發明之致免疫性胜肽。 具有高HLA-A2結合親和力之胜肽傾向具有白胺酸或 甲硫丁胺酸殘基為從N端的第二個胺基酸,與纈胺酸或白 胺酸殘基為C端胺基酸。因此,本發明也包括具有序列辨 識號:2至4之胺基酸序列的胜肽,其中所述序列辨識號 之胺基酸序列之N端的第二個胺基酸被白胺酸或甲硫丁胺 酸取代,及/或其中所述序列辨識號之胺基酸序列之C端胺 基酸被纈胺酸或白胺酸取代。可將取代引入不止於末端胺 基酸,也可於胜肽之潛在T細胞受器(TCR)辨認位置。一些 研究已證實於一具有胺基酸取代之胜肽可等於或比原來更 好,例如CAPl、p53 ( 264 - 2 72 ), Her~2/neu C 36 9 -377 ) 或 gplOO ( 209- 2 1 7 ) (Zaremba et al. Cancer Res. 57, 4570-4577, 1997, T. K. Hoffmann et al. J Immunol. (2002) Feb 1;168(3): 1338-47., S. 0. Dionne et al, Cancer Immunol immunother. (2003)52: 199-206 and S. 0. Dionne et al. Cancer Immunology, Immunotherapy (2004) 53, 307-314)。 本發明也考慮一、二個或數個胺基酸對所需胜肽之N 及/或C端的加入。本發明也包括具有高HLA抗原結合親和 力且維持細胞毒殺性T淋巴球誘發能力之此種經修飾的胜 肽。 24 201200525 然而’當胜肽序列盥—呈古Τ η 具有不同功能之内生或外生蛋 白質之胺基酸序列的一部份相 仞相同時,可能誘導副作用,例 如自體免疫疾病及/或或抗牿定私 仉将疋物質之過敏症候群。因此, 較佳為使用可得之資料庫首先執 疋執仃同源搜尋以避免胜肽之 胺基酸序列符合其他蛋白質 負之胺基酸序列的情況。當其由 與目標胜肽比較時不只存在1古 仔在具有一或兩個胺基酸不同之胜 肽的同源搜尋變得清楚時,為了增加其與慰抗原之結合 親和力,及/或增加其細胞毒殺性丁淋巴球誘發能力而不具 副作用之任何危險,可修飾目標胺基酸。 雖然如上述之具有對HLA抗原高結合親和力的胜肽被 預期為高效能,但根據作為指示之高親和表現而被選擇之 候選胜肽,更進一步被測試細胞毒殺性τ淋巴球誘發能力 的表現。此處措辭“細胞毒殺性Τ淋巴球誘發能力,,意指 虽表現於抗原呈現細胞時,胜肽誘導細胞毒殺性τ淋巴球 的能力。此外,“細胞毒殺性τ淋巴球誘發能力,,包括胜 狀誘導細胞毒殺性τ淋巴球活化、細胞毒殺性τ淋巴球增 殖、促進細胞毒殺性τ淋巴球分解目標細胞與增加細胞毒 殺性Τ淋巴球IFN- τ產生的能力。 藉由誘導攜帶人類MHC抗原之抗原呈現細胞(例如Β-淋巴球、巨噬細胞與樹突細胞(DCs))或更專一地來自人類 周邊血液單核細胞之樹突細胞,並在以胜肽刺激之後與CD8 陽性細胞混合,且之後測量由抗目標細胞之細胞毒殺性T 淋巴球產生並釋放之IFN- τ來達成細胞毒殺性T淋巴球誘 發能力的確定。如此反應系統,可使用已被產生來表現人 25 201200525 類HLA之基因轉殖動物(例如,於BenMohamed L, Kr i shnan R, Longmate J, Auge C, Low L, Primus J, Diamond DJ, Hum Immunol 2000 Aug,61 (8): 764-79 中的描述)。例 如可以51Cr與此類放射標示目標細胞,且可從自目標細胞 釋放出的放射活性計算細胞毒殺活性。或者在攜帶經固定 之胜狀的抗原呈現細胞存在下,藉由測量由細胞毒殺性τ 淋巴球產生並釋放的IFN-τ,且使用抗IFN-τ單株抗體 來使於培養基上之IFN_ r可看見來分析細胞毒殺性τ淋巴 球誘發能力。 由於如上述測試胜肽之細胞毒殺性T淋巴球誘發能 力,發現於此顯示之擇自由序列辨識號:2至4 ,特別是3 所指出的胺基酸序列中之九胜肽或十胜肽,具有特別高之 細胞毒殺性T淋巴球誘發能力與對HLA抗原之高結合親和 力。因此這些胜肽為本發明之較佳實施例。 此外,同源分析之結果顯示那些胜肽不與來自任何其 他已知人類基因產物之胜肽有顯著之同源性。當用於免疫 治療時,其降低了未知或不需要之免疫反應提升的可能 性。因此,也來自此態樣,這些胜肽提供在癌症病患中引 起抗MYBL2免疫反應的用途。因此本發明之胜肽較佳為具 有擇自序列辨識號:2至4中,特別是3之胺基酸序列的 胜肽。 除了上述本發明胜肽之修飾外,所需胜肽可被進一步 連接至其他物質,只要它們維持原始胜肽之細胞毒殺性τ 淋巴球誘發能力,且更佳為維持必不可少之HU結合。示 26 201200525 例之物質包括:胜狀、脂質、糖與糖鏈、乙醯基,天然與 合成之聚合物等。本發明胜肽可額外包含修飾,例如醣基 化支鏈氧化及/或磷酸化,只要修飾不損壞原始胜肽之生 物活性。此類修飾可授予額外之功能(例如,目標功能與 傳送功能)及/或或穩定胜肽。 例如,為了 i.77 h叩增加多胜肽之穩定度,本技術領 域已知引入D-胺基酸、胺基酸模仿物或非天然胺基酸;此 内容也適合本發明之多胜肽。可以一些方法分析一多胜肽 的穩定度。例如,可使用肽酶與多種生物培養基,例如人 類血浆與血清,來測試穩定度(參見,例如Verh〇ef M ,McFarland et al., Proc Natl Acad Sci USA 1982, 79: 6409-13). Thus, in one embodiment, the peptide of the present invention has a cytotoxic T lymphocyte evoking ability and is selected from the sequence identification numbers: 2 to 4, and the specific 21 201200525 is the amino acid sequence of 3, wherein Insert, delete and/or replace one, two or even more amino acids. It is well known to those skilled in the art that changing the individual addition or substitution of a single amino acid or a small percentage of amino acid to an amino acid sequence tends to produce the property of preserving the original amino acid branch. Thus, they are often referred to as "conservative substitutions" or "conservative modifications" in which a change in a protein results in a modified protein having a function similar to that of the original protein. The conservative representation of providing functionally similar amino acids is well known in the art. Examples of the characteristics of the amino acid bond required to be protected include, for example, hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T) and a branch having the following common functional groups or characteristics: an aliphatic branch (G, A, V, L, I, P); Branch with hydroxyl group (S, Τ, Y); branch with sulfur atom (C, M); branch with carboxylic acid and amine (D, N, E, Q); branch with alkali (R, K, H); and containing aromatic branches (H, F, Y, W). In addition, the following eight groups are each included in the art as a conservatively substituted amino acid: 1) alanine (A), glycine (G); 2) aspartic acid (D), glutamine Acid (E); 3) Aspartame (N), glutamine (Q); 4) Arginine (R), lysine (K); 5) Isoleucine (I), white Amino acid (L), thioglycolic acid (M), proline (V); 6) albinoic acid (F), tyrosine (Y), tryptophan (w); 5 201200525 7) silk Amino acid (S), sulphonic acid (Τ); and 8) cysteine (C), methylthiobutyric acid (Μ) (see, for example, Creighton, Proteins 1984). Such conservatively modified peptides are also considered to be peptides of the invention. However, the peptide of the present invention is not limited thereto, and may include a non-conservative modification as long as the resulting peptide is produced to maintain the cytotoxic T lymphocyte stimulating ability of the original peptide. Further, the modified peptide does not exclude a polymorphic variant, an interspecies homologues, and a cytotoxic τ lymphocyte-inducing peptide of the MYBL2 allele (alleles). In order to maintain the necessary cellular toxic T lymphocyte eliciting ability, a small number (e.g., one, two or several) or a small percentage of amino acid can be modified (inserted by 'deletion, addition and/or substitution). The term "several" means 5 or less amino acids, for example 4, 3 or less. The percentage of the amine group to be modified is preferably 2% or less. 15% or less 'and even more preferably, 10% or less, or 1 to 5%. Further, the peptide of the present invention may be inserted, substituted or added with an amino acid residue or an amino acid residue. Can be deleted to achieve a higher affinity. When used in the immunotherapy herein, the peptide of the present invention should be expressed on the surface of a cell or exosome, preferably as a complex with Hu antigen. Therefore, it is preferred to select a peptide which not only induces a cytotoxic thymocyte but also has a high affinity for an HLA antigen. For this purpose, (iv) can be replaced, inserted and/or added by an amino acid residue. Modification to produce a modified peptide having improved binding affinity. In addition to the naturally expressed peptide, 23 201200525 is known to bind to the peptide sequence represented by the HLA antigen (j Immunol 1994, 152: 3913; Immunogenetics 1995, 41: 178; J Immunol 1994, 155: 4307) 'So can be based on Modification of this rule introduces the immunogenic peptide of the present invention. The peptide having high HLA-A2 binding affinity tends to have a leucine or methionine residue as the second amino acid from the N-terminus, The amino acid or leucine residue is a C-terminal amino acid. Accordingly, the present invention also encompasses a peptide having the amino acid sequence of SEQ ID NO: 2 to 4, wherein the amino acid sequence of the sequence number is The second amino acid at the N-terminus is substituted with leucine or methylthiobutanoic acid, and/or the C-terminal amino acid of the amino acid sequence of the sequence number is substituted with amic acid or leucine. Substitutions can be introduced not only at the terminal amino acid, but also at the potential T cell receptor (TCR) of the peptide. Some studies have demonstrated that a peptide with an amino acid substitution can be equal to or better than the original. For example, CAP1, p53 ( 264 - 2 72 ), Her~2/neu C 36 9 -377 ) or gplOO ( 209- 2 1 7 ) (Zaremba et al. Cancer Res. 57, 4570-4577, 1997, TK Hoffmann et Al. J Immunol. (2002) Feb 1;168(3): 1338-47., S. 0. Dionne et al, Cancer Immunol immunother. (2003) 52: 199-206 and S. 0. Dio Nne et al. Cancer Immunology, Immunotherapy (2004) 53, 307-314). The invention also contemplates the addition of one, two or several amino acids to the N and/or C terminus of the desired peptide. The present invention also encompasses such modified peptides having high HLA antigen binding affinity and maintaining cytotoxic T lymphocyte eliciting ability. 24 201200525 However, when a part of the amino acid sequence of an endogenous or exogenous protein with different functions is identical, it may induce side effects, such as autoimmune diseases and/or Or anti-adhesive drugs will be allergic to the substance. Therefore, it is preferred to use the available database to first perform a homologous search to avoid the amino acid sequence of the peptide being in conformity with the amino acid sequence of the other protein. When it is compared with the target peptide, there is more than one. When the homologous search for a peptide having one or two amino acids is clear, in order to increase its binding affinity with the comfort antigen, and/or increase The cytotoxic lytic lymphocyte-inducing ability without any risk of side effects can modify the target amino acid. Although the peptide having high binding affinity for HLA antigen as described above is expected to be highly potent, the candidate peptide selected as the indicated high affinity performance is further tested for the cytotoxicity of the cell toxic lymphocytes. . The phrase "cytotoxic lymphocyte-inducing ability" means the ability of a peptide to induce a cytotoxic lymphocyte in a cell, although it is expressed in an antigen-presenting cell. In addition, "the cytotoxic lytic lymphocyte-inducing ability, including The traits induce the activation of cytotoxic lymphocytes, the proliferation of cytotoxic lymphocytes, and the ability of cytotoxic lymphocytes to decompose target cells and increase the production of cytotoxic lymphocytes IFN- τ. Presenting cells (such as sputum-lymphocytes, macrophages, and dendritic cells (DCs)) or more specifically dendritic cells derived from human peripheral blood mononuclear cells by inducing antigens carrying human MHC antigens, and After stimulation, it was mixed with CD8-positive cells, and then IFN-τ produced and released by the cytotoxic T lymphocytes of the target cells was measured to determine the cytotoxic T lymphocyte-inducing ability. Such a reaction system can use genetically modified animals that have been produced to express human 25 201200525 HLA (for example, at BenMohamed L, Kr i shnan R, Longmate J, Auge C, Low L, Primus J, Diamond DJ, Hum Immunol 2000 Aug, 61 (8): Description in 764-79). For example, 51Cr can be labeled with such a radioactive target cell, and the cytotoxic activity can be calculated from the radioactivity released from the target cell. Or by measuring IFN-τ produced and released by cytotoxic tau lymphocytes in the presence of antigen-presenting cells carrying a fixed trait, and using anti-IFN-τ monoclonal antibody to IFN_r on the medium It can be seen to analyze the cytotoxicity of the cytotoxic lymphocytes. Due to the cytotoxic T lymphocyte eliciting ability of the test peptide as described above, it was found that the free sequence identification number shown here: 2 to 4, in particular, the nine peptide or the ten peptide in the amino acid sequence indicated by 3 It has a particularly high cytotoxic T lymphocyte inducing ability and high binding affinity to HLA antigen. These peptides are therefore preferred embodiments of the invention. Furthermore, the results of the homology analysis showed that those peptides did not have significant homology to peptides derived from any other known human gene product. When used in immunotherapy, it reduces the likelihood of an unknown or unwanted immune response increase. Therefore, also from this aspect, these peptides provide the use of an anti-MYBL2 immune response in cancer patients. Therefore, the peptide of the present invention is preferably a peptide having a sequence selected from the group consisting of 2 to 4, particularly 3 amino acid sequence. In addition to the above-described modification of the peptide of the present invention, the desired peptide can be further linked to other substances as long as they maintain the cytotoxic thymocyte inducing ability of the original peptide, and more preferably maintain the essential HU binding. The substances in the example of 2012 20122525 include: victoria, lipid, sugar and sugar chain, acetyl group, natural and synthetic polymers. The peptide of the present invention may additionally comprise modifications such as glycosylation branch oxidation and/or phosphorylation as long as the modification does not damage the biological activity of the original peptide. Such modifications may confer additional functionality (eg, target function and delivery function) and/or or stabilize the peptide. For example, in order to increase the stability of a multi-peptide in i.77 h叩, it is known in the art to introduce a D-amino acid, an amino acid mimetic or an unnatural amino acid; this content is also suitable for the multi-peptide of the present invention. . The stability of a multi-peptide can be analyzed in some ways. For example, peptidase can be used to test for stability using a variety of biological media, such as human plasma and serum (see, for example, Verh〇ef M,

Eur J Drug Metab Pharmacokin 1986, 11: 29卜302)。 此外,如上所提到,在藉由一、二或數個胺基酸殘基 取代、刪除或加入之經修飾的胜肽中,可篩選或選擇與原 始胜肽相較具有相同或較高之活性的那些。因此本發明也 提供篩選或選擇與原始相較具有相同或較高之活性的經修 飾胜肽的方法。說明之方法包括下列步驟: a:將至少一個本發明胜肽之胺基酸殘基取代、刪除或 加入, b .確定胜狀的活性,與 C :選擇與原始相較具有相同或較高之活性的胜肽。 此處’要被分析之活性可包括HLA結合活性、抗原呈 現細胞或細胞毒殺性T淋巴球誘發能力與細胞毒性活性。 此處,本發明之胜肽也可被描述為“ MYBL2胜肽,,或 “MYBL2多胜肽”。 27 201200525 111. MYBL2胜肽之製備 使用熟知之技術可製備本發明之胜肽。例如,使用重 組DNA技術或化學合成可以合成方法地製備胜狀。本發明 胜肽可單獨合成或為由兩或多個胜肽所組成之較長多胜 肽。之後可分離此胜肽,即純化或分離以使其實質上無其 他自然發生之宿主細胞蛋白質與其片段或任何其他化學物 質。 本發明胜肽可包含修飾,例如醣基化、支鏈氧化或磷 酸化,其提供修飾不損壞原始胜肽之生物活性。其他修飾 包括可用來,例如增加胜肽之血清半衰期之卜胺基酸或其 他胺基酸模仿物的合併。 藉由根據經選擇之胺基酸序列的化學合成可獲得本發 明之胜肽。適合此合成之一般胜肽合成方法的例子包括, 但不限於: (i) 胜肽合成(Peptide Synthesis) Interscience, New York, 1966; (ii) 蛋白質(The Proteins), Vol. 2, Academic Press, New York, 1976; (iii) 胜肽合成(Peptide Synthesis)(in Japanese), Maruzen Co. , 1 975; (iv) 胜肽合成之基礎與實驗(Basics and Experiment of Peptide Synthesis) (in Japanese), Maruzen Co., 1 985; (v) 藥學的發展(Development of Pharmaceuticals) 28 201200525 (second volume) (in Japanese), Vo 1. 14 (peptide synthesis), Hirokawa, 1991; (vi) W099/67288 ;以及 (vii) Barany G. &amp; Merrifield R.B., Peptides Vol. 2,“Solid Phase Peptide Synthesis”,Academic Press, New York, 1980, 100-118 。 或者’藉由使用任何已知產生胜肽之基因工程方法可 獲得本發明之胜肽(例如’ M〇rris〇n j,】Bacteri〇i〇gy 1977, 132: 349-51; C1ark-Curtiss &amp; Curtiss, Methods in Enzymology (eds. Wu e纟 a/.) 1983, 101: 347-62)。 例如,首先製備一適合之載體,其懷有編碼出在一可表達 的形式中之目標胜肽的一多核苷酸(例如,對應於啟動子 序列之調控序列的下游),並將載體轉殖進入適合之宿主 細胞。之後培養宿主細胞以產生感興趣之胜肽。使用一 ^ KJ iro轉譯系統可ir〇產生胜狀。 IV.多核苷酸 本發明也提供-多核普酸,其編瑪出任何本發明上述 之胜肽。這些包括來自自然發生之舰之基因(㈤⑽ Accession No· _一〇〇2466 (序列辨識號:5))的多核普 酸與具有其之保守修飾之核苦酸序列的那些。此處措辭 保守修飾之核苦酸序列,,指序列其編石馬出相同或實質上 相同之胺基酸序列。由於基因密碼的退化,-大份之功能 相同之核酸編崎出任何已知蛋白質。例如,密碼gca、gcc、 CG與GCU皆編碼出胺基酸丙胺酸。因此,於藉由一密碼 29 201200525 具體指定丙胺酸之每個位置’可改變密碼成為任何上述不 會改變編碼出之胜狀的對應密碼。此核酸變化為“、尤默變 化(si lent variation)” ,其為保守修飾變化的一種。此 處編碼出一胜肽之每個核酸序列也描述核酸之每種可能的 沈默變化。熟悉此技藝人士明白於一核酸中各密碼(除了 AUG ’其原本為甲硫胺酸之唯一密碼、與tgg其原本為色胺 酸之唯一达、碼)可被修飾以產生一功能相同分子。因此編 碼出一胜肽之核酸的各沈默變化係為於各所揭露之序列中 被暗不性描述。 本發明之多核苷酸可由DNA、RNA與其衍生物所組成。 DNA適合地由鹼基所組成,例如鹼基a、τ、c與G,而T於 RNA中為U所取代。 本發明之多核苦酸可編碼出本發明之多個胜肽,在其 之間具有或不具有介於中間之胺基酸序列。例如介於中間 之胺基酸序列可提供多核苷酸或經轉譯之胜肽一裂解位 (例如酵素辨認序列)。更進一步而言,多核苷酸可包括 任何額外之序列至編碼出本發明胜肽之編碼序列。例如, 多核苷酸可為一重組多核苷酸,其包括胜肽表現所需之調 控序列’或可為一具有標誌基因與此類之表現載體(質 體)。一般而言’可製備此重組多核苷酸,藉由經由使用 例如聚合酶與内切酶之一般重組技術的多核苷酸操作。 可使用重組與化學合成技術兩者以產生本發明之多核 苷酸。例如,藉由插進入一適合之載體可產生一多核苷酸, 當轉染進入一勝任細胞時,其可被表現。或者,使用 30 201200525 技術或表現於適合的宿主可將一多核苷酸放大(參見,例 士口 Sambrook et al. , Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1989)。或者’使用固態技術如於BeaucageSLS IyerRP, Tetrahedron 1992, 48: 2223-311; Matthes et al., EMBO J 1984,3: 801-5中所敘述,可合成多核苷酸。 V. 外吐小體(exosomes) 本發明進一步地提供稱為外吐小體的胞間囊泡 (intracellular vesicles),其呈現形成於本發明之胜肽 與人類白血球抗原之間的複合物於其表面上。利用例如 Japanese Patent Application Kohyo Publications Nos. Hei 11_510507與W099/03499所詳述的方法以及從接受 治療和/或預防之病人所得的抗原表現細胞可製備外吐小 體。本發明之外吐小體可如疫苗般地接種,以類似於本發 明的胜肽之方式。 包含在複合物中的人類白血球抗原形式必須與需要治 療和/或預防之個體的人類白血球抗原形式相符。例如,於 日本知群中,HLA-A2,(特別是α*〇201與a*0206)為普 遍且因此對於日本人病患的治療而言為適合的。高度表現 於曰本人與高加索人之中的A2型之使用有助於獲得有效 的总果。一般在臨床上’需接受治療之病患的人類白血球 抗原形式係進行預先的研究,這可適當地選擇對特定抗原 具有高度結合親合力的胜肽或經由抗原表現具有細胞毒性 T淋巴細胞誘發性的胜肽。此外,為了獲得具有高度結合 31 201200525 親合力與細胞毒性τ淋巴細胞誘發性兩者的胜肽,可,、, j μ天 然產生之MYBL2部分胜肽的胺基酸序列為基礎,然後進^ 1、2或數個胺基酸的取代、插入及/或添加。 當對本發明外吐小體使用A2型HLA抗原時,具有序列 辨識號:2至4之任一,特別是3之序列的胜肽提供用途 VI ·抗原呈現細胞 本發明也提供經分離之抗原呈現細胞,其表現形成於 HLA抗原與本發明胜肽之間的複合物於其表面。抗原呈現 細胞可來自受到治療及/或避免之病患,且藉由其本身或與 包括本發明之胜肽、外吐小體或細胞毒殺性T淋巴球之其^ 他藥物結合可被投予如疫苗。 抗原呈現細胞並不限於特定種類之細胞,且包括樹突 細胞、蘭格罕細胞(Langerhans ce 11)、巨嗜細胞、b細胞 與活化之T細胞’已知其表現蛋白質(proteinaceous)抗原 於其細胞表面以被淋巴球所辨認。由於樹突細胞為一典型 抗原呈現細胞’其於抗原呈現細胞中具最強之細胞毒殺性 T淋巴球誘導作用’樹突細胞供給使用如本發明之抗原呈 現細胞。 例如’藉由誘導來自周邊血液單核細胞之樹突細胞與 之後J λ H h 或h叩以本發明胜肽接觸(刺 激)其可獲得本發明抗原呈現細胞。當本發明之胜肽投予 至一個體’於個體身體内誘導表現本發明胜肽之抗原呈現 細胞。因此’藉由在將本發明胜肽投予至一個體後,自此 個體收集抗原呈現細胞可獲得本發明之抗原呈現細胞。或 32 ⑤ 201200525 現細胞與本發明胜肽接觸 者’藉由將自個體收集之抗原呈 可獲得本發明之抗原呈現細胞。 可將本發明之抗原呈現細胞單獨或結合包括本發明胜 肽、外吐小體或細胞毒殺性τ淋巴球的其他藥物來投予至 一個體以誘導於個體中之抗癌免疫反應。例如^ mm 投予可包括步驟: a :自一第一個體收集抗原呈現細胞, b :以胜肽接觸步驟a之抗原呈現細胞,以及 c :將步驟b之抗原呈現細胞投予一第二個體。 第個體與第一個體可為相同個體或可為不同個體。 自步驟b獲得之抗原呈現細胞可作為疫苗被投予以治療及 /或預防癌症,癌症包括睪丸腫瘤、胰臟癌、膀胱癌、非小 細胞肺癌、小細胞肺癌與食道癌。 本發明也提供製造包括抗原呈現細胞之藥學組合物的 一方法或製程,其中方法包括將本發明胜肽與藥學上可接 受之載體一起混合或配製的步驟。 根據本發明一方面,抗原呈現細胞具高程度細胞毒殺 性T淋巴球誘發能力。在用語“高程度細胞毒殺性τ淋巴 球誘發能力”中,高程度相對於藉由抗原呈現細胞沒有與 胜肽接觸或與無法誘導細胞毒殺性τ淋巴球之胜肽接觸的 程度。藉由包括/yj W 將編碼出本發明胜肽之多核苷酸 轉移至抗原呈現細胞的步驟的方法與上述之方法,可製備 此種具高程度細胞毒殺性τ淋巴球誘發能力之抗原呈現細 胞。此經引入之基因可為DNA或RNA形式。引入方法的例 33 201200525 子包括’並無特別限制’各種於此領域一般被執行的方法, 例如月曰質體轉染(lip0fecti〇n)、電穿孔法 (electroporation)與磷酸鈣方法。更特別地,可執行其如 Cancer Res 1996, 56: 5672-7; J Immunol 1998, 161: 5607-13; J Exp Med 1996, 184: 465-72; PublishedEur J Drug Metab Pharmacokin 1986, 11: 29 Bu 302). Furthermore, as mentioned above, in a modified peptide substituted, deleted or added by one, two or several amino acid residues, the same or higher than the original peptide can be screened or selected. Those active. The invention therefore also provides a method of screening or selecting a modified peptide having the same or higher activity as the original. The method comprises the steps of: a: substituting, deleting or adding at least one amino acid residue of the peptide of the present invention, b. determining the activity of the winning form, and C: selecting the same or higher than the original one. Active peptide. Here, the activity to be analyzed may include HLA binding activity, antigen presenting cells or cytotoxic T lymphocyte inducing ability and cytotoxic activity. Here, the peptide of the present invention can also be described as "MYBL2 peptide, or "MYBL2 multipeptide". 27 201200525 111. Preparation of MYBL2 peptide The peptide of the present invention can be prepared using well-known techniques. For example, The winning form can be synthesized synthetically using recombinant DNA technology or chemical synthesis. The peptide of the present invention can be synthesized alone or as a longer multi-peptide consisting of two or more peptides. The peptide can be isolated, ie purified or Is isolated so that it is substantially free of other naturally occurring host cell proteins and fragments thereof or any other chemical. The peptides of the invention may comprise modifications, such as glycosylation, branched oxidation or phosphorylation, which provide modification without damaging the original peptide Biological activity. Other modifications include combinations of amino-amino acids or other amino acid mimetics that can be used, for example, to increase the serum half-life of the peptide. The invention can be obtained by chemical synthesis according to selected amino acid sequences. Peptides. Examples of general peptide synthesis methods suitable for this synthesis include, but are not limited to: (i) Peptide Synthesis Interscience, New York, 1966; (ii) Protein (The Proteins), Vol. 2, Academic Press, New York, 1976; (iii) Peptide Synthesis (in Japanese), Maruzen Co., 1 975; (iv) Basis and experiment of peptide synthesis (Basics and Experiment of Peptide Synthesis) (in Japanese), Maruzen Co., 1 985; (v) Development of Pharmaceuticals 28 201200525 (second volume) (in Japanese), Vo 1. 14 (peptide synthesis) , Hirokawa, 1991; (vi) W099/67288; and (vii) Barany G. &amp; Merrifield RB, Peptides Vol. 2, "Solid Phase Peptide Synthesis", Academic Press, New York, 1980, 100-118. The peptide of the present invention can be obtained by using any genetic engineering method known to produce a peptide (for example, 'M〇rris〇nj, 】 Bacteri〇i〇gy 1977, 132: 349-51; C1ark-Curtiss &amp; Curtiss, Methods in Enzymology (eds. Wu e纟a/.) 1983, 101: 347-62). For example, a suitable vector is first prepared which harbors a polynucleotide encoding a target peptide in an expressible form (eg, downstream of a regulatory sequence corresponding to a promoter sequence) and transfers the vector Colonization into a suitable host cell. The host cells are then cultured to produce a peptide of interest. Using a ^ KJ iro translation system can generate a victory. IV. Polynucleotides The present invention also provides a multi-nucleotide acid which encodes any of the above-described peptides of the present invention. These include those from the genes of naturally occurring ships ((5) (10) Accession No. _2466 (SEQ ID NO: 5)) and those with conservatively modified nucleotide sequences. The term "conservatively modified nucleotide acid sequence" as used herein refers to a sequence of amino acid sequences which are identical or substantially identical. Due to the degradation of the genetic code, the large number of nucleic acids with the same function encodes any known protein. For example, the codes gca, gcc, CG, and GCU all encode amino acid alanine. Therefore, the password can be changed by specifying a position of each of the alanine by a password 29 201200525 to become any corresponding password which does not change the encoded winning. This nucleic acid change is ", sir lent variation", which is one of the conservative modification changes. Each nucleic acid sequence encoding a peptide here also describes every possible silent change in the nucleic acid. Those skilled in the art will appreciate that the various codes in a nucleic acid (except AUG' which is originally the unique code for methionine, and the unique code for tgg which is originally a tryptophan) can be modified to produce a functionally identical molecule. Thus, each silent change in the nucleic acid encoding a peptide is described as being implicitly described in each of the disclosed sequences. The polynucleotide of the present invention may be composed of DNA, RNA and derivatives thereof. The DNA suitably consists of bases such as bases a, τ, c and G, and T is substituted by U in RNA. The polynucleic acid of the present invention encodes a plurality of peptides of the present invention with or without an intermediate amino acid sequence therebetween. For example, an intermediate amino acid sequence can provide a polynucleotide or a translated peptide-cleavage site (e.g., an enzyme recognition sequence). Still further, a polynucleotide may comprise any additional sequence to a coding sequence that encodes a peptide of the invention. For example, the polynucleotide may be a recombinant polynucleotide comprising a regulatory sequence required for the performance of the peptide or may be a representation vector (plastid) having a marker gene and such. Generally, such recombinant polynucleotides can be prepared by manipulation via a polynucleotide using a general recombinant technique such as a polymerase and an endonuclease. Both recombinant and chemical synthesis techniques can be used to produce the polynucleotides of the invention. For example, a polynucleotide can be produced by insertion into a suitable vector which can be expressed when transfected into a competent cell. Alternatively, a polynucleotide can be amplified using the technique of 30 201200525 or expressed in a suitable host (see, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1989). Alternatively, polynucleotides can be synthesized using solid state techniques as described in Beaucage SLS Iyer RP, Tetrahedron 1992, 48: 2223-311; Matthes et al., EMBO J 1984, 3: 801-5. V. Exosomes The present invention further provides intercellular vesicles called exocytosis, which exhibit a complex formed between the peptide of the present invention and human leukocyte antigen. On the surface. Exosomes can be prepared using methods such as those detailed in Japanese Patent Application Kohyo Publications Nos. Hei 11_510507 and WO99/03499, and antigen-expressing cells obtained from patients receiving treatment and/or prevention. The exosome of the present invention can be vaccinated as a vaccine in a manner similar to the peptide of the present invention. The human leukocyte antigen form contained in the complex must conform to the human leukocyte antigen form of the individual in need of treatment and/or prevention. For example, in the Japanese group, HLA-A2, (especially α*〇201 and a*0206) is common and thus suitable for the treatment of Japanese patients. High performance The use of the A2 type between the person and the Caucasian helps to obtain an effective total fruit. It is generally pre-study in the clinical form of human leukocyte antigens in patients who are in need of treatment, which can appropriately select peptides with high binding affinity for specific antigens or cytotoxic T lymphocyte-induced properties via antigens. The peptide. In addition, in order to obtain a peptide with high binding 31 201200525 affinity and cytotoxic tau lymphocyte-inducing, it is possible to, based on, the amino acid sequence of the naturally occurring MYBL2 partial peptide of j μ, and then enter Substitution, insertion and/or addition of 2 or several amino acids. When the A2 type HLA antigen is used for the exosome of the present invention, the peptide having the sequence identification number: 2 to 4, particularly the sequence of 3, provides use VI. Antigen presenting cells The present invention also provides isolated antigen presentation. A cell which exhibits a complex formed between the HLA antigen and the peptide of the present invention on its surface. The antigen presenting cells may be derived from a patient being treated and/or avoided and may be administered by itself or in combination with a drug comprising the peptide of the present invention, exosome or cytotoxic T lymphocytes. Such as a vaccine. Antigen presenting cells are not limited to a particular type of cell, and include dendritic cells, Langerhans cells (Langerhans ce 11), macrophages, b cells, and activated T cells, which are known to represent proteinaceous antigens. The cell surface is recognized by the lymphocytes. Since dendritic cells are a typical antigen-presenting cell, which has the strongest cytotoxic T lymphocyte-inducing effect in antigen-presenting cells, dendritic cells are supplied using antigen-presenting cells such as the present invention. For example, the antigen presenting cells of the present invention can be obtained by inducing dendritic cells derived from peripheral blood mononuclear cells to be contacted (stimulated) with the peptide of the present invention by J λ H h or h叩. When the peptide of the present invention is administered to a body, an antigen-presenting cell which exhibits the peptide of the present invention is induced in the body of the individual. Therefore, the antigen-presenting cells of the present invention can be obtained by collecting antigen-presenting cells from the individual after administration of the peptide of the present invention to a single body. Or 32 5 201200525 The present cells are contacted with the peptide of the present invention. The antigen presenting cells of the present invention can be obtained by expressing the antigen collected from the individual. The antigen-presenting cells of the present invention can be administered to a single body to induce an anti-cancer immune response in an individual, alone or in combination with other drugs including the peptide of the present invention, exosome or cytotoxic lymphocytes. For example, ^mm administration can include the steps of: a: collecting antigen presenting cells from a first individual, b: presenting the cells with the peptide contacting step a, and c: administering the antigen presenting cells of step b to a second individual . The first individual and the first individual may be the same individual or may be different individuals. The antigen-presenting cells obtained from the step b can be administered as a vaccine for treating and/or preventing cancer, and the cancer includes a testicular tumor, a pancreatic cancer, a bladder cancer, a non-small cell lung cancer, a small cell lung cancer, and an esophageal cancer. The invention also provides a method or process for making a pharmaceutical composition comprising an antigen presenting cell, wherein the method comprises the step of mixing or formulating the peptide of the invention with a pharmaceutically acceptable carrier. According to one aspect of the invention, the antigen presenting cells have a high degree of cytotoxic T lymphocyte evoking ability. In the phrase "high degree of cytotoxic tauria-inducing ability", the degree of contact with the peptide by the antigen-presenting cells or the peptide which is incapable of inducing the cytotoxic tau lymphocytes is highly correlated. The antigen-presenting cell having the high degree of cytotoxic lytic lymphocyte inducing ability can be prepared by the method comprising the step of transferring the polynucleotide encoding the peptide of the present invention to the antigen-presenting cell by the method of /yj W and the method described above. . The introduced gene can be in the form of DNA or RNA. Examples of introduction methods 33 201200525 Sub-includes 'are not particularly limited' various methods generally performed in this field, such as scorpion plastid transfection (lip0fecti〇n), electroporation (electroporation) and calcium phosphate method. More particularly, it can be performed as in Cancer Res 1996, 56: 5672-7; J Immunol 1998, 161: 5607-13; J Exp Med 1996, 184: 465-72;

Japanese Translation of International Publication No. 20 00-509281中所述。藉由轉移基因進入抗原呈現細胞, 基因遭遇轉錄、轉譯與此類於細胞中,且之後處理所獲得 之蛋白質,並經由一呈現途徑來進行以與呈現本發明胜肽。 V11 ·細胞毒殺性τ淋巴球 抗任何本發明胜肽誘導之細胞毒殺性τ淋巴球增強“ η叩以癌症細胞為標的之免疫反應,且因此就其本身而 言,可被使用為疫苗以相似於胜肽之方式。因此本發明也 提供經分離之細胞毒殺性Τ淋巴球其藉由任何本發明之胜 肽專一地被誘導或活化。 可獲得此種細胞毒殺性7淋巴球,藉由⑴將本發明 胜肽投予至-個體’從此個體收集細胞毒殺性τ淋巴球; 或(2 );!字來自個體之抗原呈現細胞與⑽陽性細胞或周遠 血液單核淋巴球與本發明之胜肽以&gt;〇接觸(刺激)且 之後分離細胞毒殺性Τ壯p 。^ 丨1林巴球,或(3)將CD8陽性細胞或Japanese Translation of International Publication No. 20 00-509281. By transferring the gene into the antigen-presenting cell, the gene undergoes transcription, translation and such in the cell, and then the obtained protein is processed and carried out via a presentation pathway to present the peptide of the present invention. V11 - cytotoxic tau lymphocytes against any of the peptide-inducible cytotoxic tau lymphocytes of the present invention enhances the immune response of η叩 to cancer cells, and thus, as such, can be used as a vaccine similarly The present invention also provides an isolated cytotoxic axillary lymphocyte which is specifically induced or activated by any of the peptides of the present invention. Such a cytotoxic 7 lymphocyte can be obtained by (1) The peptide of the present invention is administered to an individual to collect a cytotoxic tau lymphocyte from the individual; or (2); the word is derived from an individual antigen presenting cell and (10) positive cell or a peripheral blood mononuclear lymphocyte and the present invention The peptide is contacted (stimulated) with &gt; 且 and then isolated by cytotoxicity p ^ ^ 林 1 linba ball, or (3) CD8 positive cells or

周邊血液單核淋巴球與矣規A 八兴衣現HLA抗原與胜肽之複合物於其 表面上之抗原呈現細胞或外吐小體以心接觸且之後分 離細胞毒殺性T淋巴玻·劣(j ^ ?丨 朴G坏,或(4)引入包括編碼出與與本發 明胜狀結合之T細月命夸-A g α办 也又體-人早TL的多核苷酸的基因。藉由 34 201200525 上述方法可製帛i述抗原呈現 法被詳細敘述於下方“viii 中。 細胞或外吐小體,且(4)之方 T細胞受體(TCR),,的段落 本發明細胞毒殺性Τ淋巴球 《 避免之病患,且可藉由$到治療及/或 發明*由”本身或為了調節作用而與包括本 發月之胜肽或之外吐小體的其他藥物結合來被投予。所獲 仲之細胞毒殺性[淋巴球起專一抗目標細胞的作用,而目 標細胞其表現本發明胜肽,例如用於誘導之相同胜肽。目 標細胞可為細胞其内生性表%咖,例如癌細胞,或被 以#㈣基因轉殖之細胞;且由於藉由胜肽刺激表現本發 明胜肽於細胞表面之細胞,也可做為經活化之細胞毒殺性 T淋巴球攻擊的目標。 VI11_ τ細胞受體(TCR) 本發明也提供一組合物其包含由編碼出可形成τ細胞 受體之次單位之多胜肽的核酸,與其使用方法。Τ細胞受 體之次單位具有能力形成τ細胞受體,其授與專一性至抗 腫瘤細胞的Τ細胞’腫瘤細胞表現MYBL2。藉由使用本技 術領域所知的方法可確認作為細胞毒殺性Τ淋巴球之τ細 胞受體次單元之〇:-與石—支鏈的核酸,而細胞毒殺性τ淋 巴球以一或多個本發明之胜肽所誘導(w〇2〇〇7/〇32255與Peripheral blood mononuclear lymphocytes and sputum A octopus complex HLA antigen and peptide complexes on the surface of the antigen present cells or exocytosis in the heart and then separate the cell toxic T lymphocyte j ^ 丨 G G G bad, or (4) the introduction of a gene encoding a polynucleotide which is combined with the singularity of the present invention and which is combined with the singularity of the invention. 34 201200525 The above method can be described in detail in the following "viii. Cell or exosome, and (4) square T cell receptor (TCR), paragraph of the present invention cell cytotoxicity A lymphocyte "avoiding a disease, and can be administered by itself to a therapeutic or/or invention*, or for the purpose of modulation, in combination with other drugs including the peptide of the present month or the exosome The obtained cytotoxicity of the secondary cells [the lymphocytes act exclusively against the target cells, and the target cells exhibit the peptides of the present invention, for example, the same peptides for induction. The target cells may be the endogenous tables of the cells. Such as cancer cells, or cells that have been transferred to the #(四) gene; The peptide stimulates the expression of the peptide of the present invention on the cell surface, and can also serve as a target for activated cytotoxic T lymphocyte challenge. VI11_τ cell receptor (TCR) The present invention also provides a composition comprising the coding A nucleic acid capable of forming a multi-peptide of a subunit of a tau cell receptor, and a method of using the same, the subunit of the sputum cell receptor has the ability to form a tau cell receptor, which confers a specific cell to a tumor cell of a tumor cell The cells express MYBL 2. The τ cell-receptor subunits of the cytotoxic lymphocytes are identified by using methods known in the art: - and stone-branched nucleic acids, while the cytotoxic tau lymphocytes are Induction of one or more peptides of the invention (w〇2〇〇7/〇32255 with

Morgan et al.,J immun〇i,171,3288 (2003))。例如, 喜好以聚合酶鏈鎖反應方法來分析T細胞受體次單元。用 於分析之聚合酶鏈鎖反應引子可為,例如5’ -R引子 (5, -gtctaccaggcattcgcttcat-3’ )為 5’ 端引子(序 35 201200525 列 辨識號 : 7 ) 與 3-TRa-C 引 子 (5’ -tcagctggaccacagccgcagcgt-3’ )專一於 τ 細胞受 體alpha鏈C區(序列辨識號:8) 、3-TRb-Cl引子 (5’ -tcagaaatcctttctcttgac-3’ )專一於 τ 細胞受體 beta鍵C1區(序列辨識號:9)或3-TRbeta-C2引子(5,~ ctagcctctggaatcctttctc1;t-3’ )專一於 T 細胞受體 beta 鏈C2區(序列辨識號:10)為3’ 端引子,但不限於其。 引出之T細胞受體可以高親合力結合表現MYBL2胜肽之目 標細胞,且視需要/y? f/fo與“ h iro居中有效殺死表現 MYBL2胜肽之目標細胞。 編碼出T細胞受體次單位的核酸序列可合併進入適合 之載體,例如反轉錄病毒載體。這些載體為本技術領域所 熟知。通常包含其之核酸或載體可被轉移至一 T細胞,例 如一來自一病患之T細胞。有用地,本發明提供一現成 (of f-the-shel f)的組合物允許快速修飾病人所擁有之τ 細胞(或其他哺乳動物之那些)以快速簡單產生具有優秀 之癌症細胞殺死特性的經修飾Τ細胞。 特定之Τ細胞受體可專一地辨認本發明之一胜肽與 HLA分子之複合物,當Τ細胞受體於Τ細胞表面時,給予Τ 細胞抗目標細胞之專一活性。藉由任何已知方法可確認上 述複合物之專一辨認’而較佳方法包括,例如使用HLA分 子與本發明胜肽之四聚體(tetramer)分析,與ELI SPOT分 析。藉由執行EL I SPOT分析,其可確認表現τ細胞受體於 細胞表面上之T細胞藉由T細胞受體辨認一細胞,且訊息 ⑤ 201200525 傳达於細胞内。當複合物存在於τ細胞表面時藉由已知方 法也可執行上述複合物可給予一 τ細胞細胞毒性活性的確 認。較佳方法包括,你丨‘,&gt; UT Λ 例如’抗HLA險性目標細胞之細胞毒 性活性測定,例如鉻(chromium)釋放分析。 本發明也提供細胞毒殺性τ淋巴球,其藉由以編碼出 在HLA-2存在下與例如序列辨識號:2至4,特別是3之 MYBL2胜肽結合的Τ細胞受體次單位多胜肽的核酸來轉導 來製備。經轉導之細胞毒殺性Τ淋巴球可“&gt;〇引導至 癌症細胞,且可藉由熟知的培養方法擴張(例如Morgan et al., Jimmun〇i, 171, 3288 (2003)). For example, it is preferred to analyze the T cell receptor subunit by the polymerase chain reaction method. The polymerase chain reaction primer for analysis can be, for example, a 5'-R primer (5, -gtctaccaggcattcgcttcat-3') is a 5' end primer (Sequence 35 201200525 column identification number: 7) and 3-TRa-C primer (5'-tcagctggaccacagccgcagcgt-3') specific to the t-cell receptor alpha chain C region (SEQ ID NO: 8), 3-TRb-Cl primer (5'-tcagaaatcctttctcttgac-3') specific to tau cell receptor beta bond The C1 region (SEQ ID NO: 9) or the 3-TRbeta-C2 primer (5, ~ ctagcctctggaatcctttctc1; t-3') is specific to the T cell receptor beta chain C2 region (SEQ ID NO: 10) is the 3' primer. But not limited to it. The extracted T cell receptor can bind to the target cell expressing MYBL2 peptide with high affinity, and if necessary, /y?f/fo and "h iro centering effectively kill target cells expressing MYBL2 peptide. Encoding T cell receptor The subunit nucleic acid sequences can be combined into a suitable vector, such as a retroviral vector. Such vectors are well known in the art. Nucleic acids or vectors comprising the same can be transferred to a T cell, such as a T from a patient. Usefully, the present invention provides a ready-made (of-f-the-shelf) composition that allows rapid modification of tau cells (or other mammalian ones) possessed by a patient to quickly and easily produce excellent cancer cell killing Characteristic modified sputum cells. The specific sputum cell receptor can specifically recognize the complex of one peptide and HLA molecule of the present invention, and the specific activity of the sputum cell against the target cell when the sputum cell receptor is on the surface of the sputum cell. The specific recognition of the above complex can be confirmed by any known method, and preferred methods include, for example, the use of HLA molecules and tetramer analysis of the peptide of the present invention, and E LI SPOT analysis. By performing an EL I SPOT analysis, it was confirmed that a T cell expressing a tau cell receptor on the cell surface recognizes a cell by a T cell receptor, and message 5 201200525 is transmitted inside the cell. The above complex can also be used to confirm the cytotoxic activity of a tau cell by the known method when present on the surface of the tau cell. The preferred method includes, for example, '丨', &gt; UT Λ, for example, 'anti-HLA risk target cell Cytotoxic activity assays, such as chromium release assays. The invention also provides cytotoxic tau lymphocytes by encoding in the presence of HLA-2 with, for example, sequence identification numbers: 2 to 4, particularly 3 The MYBL2 peptide binds to the nucleic acid of the sputum cell receptor subunit polypeptide to be transduced. The transduced cytotoxic sputum lymphocytes can be "&gt; 〇 guided to cancer cells, and can be cultured by well-known methods. Expansion (for example

KaWakami J 1随unol.,142, 3452-3461 (1 989))。 本發明細胞毒殺性T淋巴球也可用來形成一致免疫組合 物’其於-需要治療或保護之病患中治療或避免癌症為有 效(W02006/031221) 〇 IX.藥學試劑或組合物 由於與正常組織相較,MYBL2表現於癌症中特別被提 高,癌症包括睪丸腫瘤、胰臟癌、膀胱癌、非小細胞肺癌、 小細胞肺癌與食道癌’本發明之胜肽或編石馬出此類胜狀之 多核苦酸可用於癌症或腫瘤之治療及/或預防,及/或避免 其手術後之復發。因此,本發明提供—藥學試劑或組合物. 用來療及/或預防癌症或腫瘤’及/或避免其手術後之復 發’此類試劑或組合物包括一或多個本發明胜狀或編碼出 此胜狀之多核普酸作為活性成分。或者,本發明之胜肽可 表現於任何鈾述外吐小體或細胞表面’例如抗原呈現細 胞,以用來作為藥學試劑或組合物。此外,上述以本發明 37 201200525 任:可胜肽為標的之細胞毒殺性τ淋巴球也可用來作為本發 明藥學試劑或組合物之活性成分。 在另-實施例中,本發明也在製造用以治療癌症或腫 瘤之藥學組合物或試劑中提供—活性成分的使用,其擇自: (a )本發明胜肽; (b)於一可表現之形式,編碼出如此處揭露之此種胜 肽的核酸; (C)表現本發明一胜肽於裒矣;u σ 职Κ具表面上之抗原呈現細胞或 外吐小體;以及 (d)本發明之細胞毒殺性了淋巴球。 或者,本發明更提供-用以治療遽瘤之癌症的活性成 分擇自: (a)本發明胜肽; ⑻於-可表現之形式,編碼出如此處揭露之此種胜 肽的核酸; (c)表現本發明一胜妝於甘生 L L , 肽於其表面上之抗原呈現細胞或 外吐小體;以及 ⑷本發明之細胞毒殺性τ淋巴球。 或者’本發明更提供—製;生田 表&amp;用以治療癌症或腫瘤之藥 學組合物或試劑的方法或製 、 茲與μ斗、‘油 裂耘,其中方法或製程包括將一 藥子上或生理上可接受之截辦伽 載體與—活性成分一起配製的步 驟,活性成分擇自: / (a) 本發明胜肽; (b) 於一可表現之形 x ’蝙碼出如此處揭露之此種胜 ⑤ 38 201200525 肽的核酸; (C )表現本發明一 外吐小體;以及 胜狀於其表面 上之抗原呈現細胞 或 (d)本發明之細胞毒殺性τ淋巴球 為活性成分。 在另-實施例中,本發明也提供一製造用以治療或避 免癌症或腫瘤之藥學组合物或試劑的方法或製程,並中方 法或製程包括將-活性成分與—藥學上或生理上可接受之 載體一起混合的步驟,其中活性成分擇自: (a) 本發明胜肽; (b) 於-可表現之形式,編碼出如此處揭露之此種胜 肽的核酸; (c) 表現本發明一胜肽於其表面上之抗原呈現細胞或 外吐小體;以及 (d) 本發明之細胞毒殺性τ淋巴球。 或者本發明藥學組合物或試劑可被用於癌症或腫瘤 之預防與其手術後之復發的避免之任一或兩者。 本發明之藥學試劑或組合物提供使用如一疫苗。在本 毛月内文中’措辭“疫苗,,(也指一致免疫組合物)意指 、’ /、精由接種至動物具有誘導抗腫瘤免疫力的功能。 本發明之藥學試劑或組合物可用於治療及/或避免癌 症或腫痼,;β 4 及/或其手術後復發的避免於一個體或病患令, 個體或,忘$ h / 匕括人類與任何其他哺乳動物,其包括,但不 限於小鼠、士 ΰ 乳、天竺鼠、兔子、雜、狗、、绵羊、山羊、 39 201200525 緒牛.馬#子、狒狒與黑獲獲,特別是一商業上重要 動物或被馴養了的動物。 根據本發明,已發現具有序列辨識號:2至4之任一 個,特別是3之胺基酸序列的胜肽為hu_a2限制之抗原決 定位胜肽或候選物,其可誘導強而專一之免疫反應。因此 包括任-具有序列辨識號:2至4 ’特別是3之胺基酸序列 之這二胜肽的本發明藥學試劑或組合物特別適合投予Η&quot; 抗原為HLA-A2之個體。相同的東西提供至包含編碼出任何 le些胜肽之多核苷酸(即,本發明之多核苷酸)的藥學試 劑或組合物。 由本發明藥學試劑或組合物治療之癌症或腫瘤不限於 且包括其中關於MYBL2 (例如,為過度表現)之所有種類 之癌症或腫瘤,包括’例如睪丸腫瘤、胰臟癌、膀胱癌、 非小細胞肺癌、小細胞肺癌與食道癌。 本發明藥學試劑或組合物可包括除了上述活性成分 外具有誘導細胞毒知·性τ淋巴球抗似癌細胞之能力的其 他胜肽、編碼出此其他胜肽之其他多核苷酸、其他表現此 其他胜肽之細胞或此類。於此,具有誘導細胞毒殺性T淋 巴球抗似癌細胞之能力的其他胜肽由癌症專一抗原所例示 (例如,經定義之腫瘤相關抗原),但不限於此。 若需要,本發明之藥學試劑或組合物可視需要包括其 他治療物質為一活性成分,只要此物質不抑制活性成分之 抗腫瘤功效,活性成分例如任何本發明胜肽。例如,配方 可包括抗發炎試劑、止痛劑、化學治療與其類似。除了包 ⑤ 40 201200525 括其他治療物質於藥劑其本身中,也可將本發明之藥劑可 與或多個其他藥學試劑相繼或同時投予。藥劑與藥學試 劑的量依照,例如使用何種藥學試劑、要治療之疾病與投 藥的計晝與方式。 應瞭解的是,除了此處特別提及之成分外,本發明之 藥學試劑或組合物可包括本技術領域一般之其他試劑,其 具有關於討論中之配方形式。 在本發明一實施例中,本發明之藥學試劑或組合物可 被包含於製造之商品與套組,其包含對於要被治療之疾 病,例如癌症的病理情況有用之材料。製造之商品可包括 具有一標籤之任何本發明藥學試劑或組合物的容器。適合 的容器包括瓿、小瓶(vial)與試管。容器可形成自各種材 料,例如玻璃或《。於容器上之標籤需指出試劑為用來 治療或避免疾病之-或多個情況^標籤也可指出投藥指示 等。 ”曰y、 、除了上述容器外,套組包括本發明藥學試劑或組合物 可視需要更進一步包括一第二容器’其儲藏一藥學上可接 受之稀釋液。其可更包括商業或使用者觀點需要之其他材 料’包括其他緩衝溶液、稀釋液、遽器、針、注射器與具 有使用說明之包裝插入物。 ° 〃 藥學試劑或組合物若需要可被呈現於-包(Pack)或一 分配器’其可包含含有活性成分之一或多單位劑量形式。 包裝可例如包括金屬或_,例如一泡棉箱⑽此 pack)。包或分配器可伴隨著投藥指示。 41 201200525 (1)藥學試劑或組合物包含胜肽作為活性成分 可直接投予本發明胜肽為一藥學試劑或組合物,若需 要的話,其可被一般配方方法所配製。在之後的例子,除 了本發明胜肽外、若適合可包括載體、賦形劑與原始做為 藥物使用之此類而無特別限制。上述載體的例子為滅菌水 生理食鹽水、磷酸緩衝溶液與培養液體(culture fluid) 與此類。更進一步而言,若必須,藥學試劑或組合物可含 安定劑、懸液劑、防腐劑、界面活性劑與此類。本發明之 藥學試劑或組合物可用來抗癌目的。 可將本發明之胜肽製備為一組合,其由兩或更多個本 發明之胜肽所組成’以h W誘導細胞毒殺性T淋巴球。 胜肽組合可以雞尾酒形式執行或可使用標準技術彼此結 合。例如’胜肽可被化學連接或表現如一單一融合多胜肽 序歹!結合之胜狀可為相同或不同。藉由投予本發明之胜 狀’藉由HLA抗原’高密度呈現胜肽於抗原呈現細胞上, 之後誘導對形成於呈現胜肽與HLA抗原之間的複合物專一 反應之細胞毒殺性T淋巴球《或者,抗原呈現細胞(例如, 樹突細胞)自一個體移出且之後以本發明胜肽刺激以獲得 表現任何本發明之胜肽於其細胞表面上之抗原呈現細胞。 將這些杬原呈現細胞再投予至該個體以於該個體中誘導細 胞毒奴性T淋巴球,且因此可增加朝向腫瘤相關内皮的侵 犯。 ~療及/或避免癌症或腫瘤之藥學試劑或組合物,其包 本發明之胜肽為活性成分,也可包含一已知為有效建 42 201200525 立細胞免疫力之佐劑。或者藥學試劑或組合物可與其他活 性成分-起被投予,或以配製成細粒被投予。佐劑指一化 合物,當與具有免疫活性之蛋白質一起投予(或依次)時, 其增強抗蛋白質之免疫反應。於此考慮之佐劑包括於文 獻(Clin M1Cr〇b1〇1 Rev 1 994, 7: m 一 89)中所描述的那 些。適合之佐劑的例子包括磷酸鋁、氫氧化鋁、明礬、霍 亂毒素、沙門氏菌毒素、佛氏不完全佐劑(Inc〇mpiete Freund*s adjuvant, 〖FA)、佛氏完全佐劑(c〇mpleteKaWakami J 1 with unol., 142, 3452-3461 (1 989)). The cytotoxic T lymphocytes of the present invention can also be used to form a consensus immunological composition which is effective in treating or avoiding cancer in a patient in need of treatment or protection (W02006/031221). IX. Pharmaceutical agents or compositions due to normal Compared with tissues, MYBL2 is particularly enhanced in cancer, including testicular tumors, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. 'The peptide of the present invention or the stone of the invention is such a victory. Polynuclear acid can be used for the treatment and/or prevention of cancer or tumors, and/or to avoid recurrence after surgery. Accordingly, the present invention provides a pharmaceutical agent or composition for treating and/or preventing cancer or tumor 'and/or avoiding recurrence after surgery'. Such agents or compositions include one or more of the present invention. This triumphant multi-nucleotide is used as an active ingredient. Alternatively, the peptide of the present invention can be expressed on any uranium exosome or cell surface&apos;, e.g., antigen presenting cells, for use as a pharmaceutical agent or composition. Further, the above-mentioned cytotoxic tau lymphocytes which are the target of the present invention 37 201200525: the peptide can also be used as an active ingredient of the pharmaceutical agent or composition of the present invention. In another embodiment, the invention also provides for the use of an active ingredient in the manufacture of a pharmaceutical composition or agent for treating cancer or a tumor, selected from: (a) a peptide of the invention; a form of expression encoding a nucleic acid of such a peptide as disclosed herein; (C) exhibiting a peptide of the present invention on a sputum; an antigen present on the surface of a u σ job having a cell or an exosome; and (d) The cells of the invention are toxic to lymphocytes. Alternatively, the present invention further provides that the active ingredient for treating cancer of the tumor is selected from: (a) a peptide of the invention; (8) a nucleic acid encoding the peptide as disclosed herein in a form that can be expressed; c) exhibiting a win of the present invention in Gansheng LL, the antigen on the surface of the peptide exhibiting cells or exocytosis; and (4) the cytotoxic tau lymphocytes of the present invention. Or 'the invention further provides a method; a method and system for treating a pharmaceutical composition or a reagent for cancer or a tumor, and a sputum, a 'cracking sputum, wherein the method or process includes placing a drug on the drug Or a physiologically acceptable step of formulating the gamma carrier together with the active ingredient, the active ingredient being selected from: / (a) the peptide of the invention; (b) in the form of a representable x' bat code as disclosed herein Such a nucleic acid of the peptide 5 38 201200525; (C) exhibiting an exosome of the present invention; and presenting an antigen presenting cell on the surface thereof or (d) the cell toxic lymphocyte of the present invention is an active ingredient . In still other embodiments, the invention also provides a method or process for making a pharmaceutical composition or agent for treating or avoiding cancer or a tumor, and wherein the method or process comprises - combining the active ingredient with - pharmaceutically or physiologically The step of admixing the accepted carriers, wherein the active ingredient is selected from: (a) a peptide of the invention; (b) a nucleic acid encoding the peptide as disclosed herein in a form that can be expressed; (c) a performance An antigen presenting a peptide on its surface exhibits a cell or an exocytosis; and (d) a cytotoxic tau lymphocyte of the invention. Alternatively, the pharmaceutical composition or agent of the present invention can be used for either or both of the prevention of cancer or tumor and the avoidance of recurrence after surgery. The pharmaceutical agent or composition of the present invention provides for use as a vaccine. In the context of this term, the wording "vaccine", (also referred to as a consensus immunological composition) means that the inoculum to the animal has the function of inducing anti-tumor immunity. The pharmaceutical agent or composition of the invention can be used for Treating and / or avoiding cancer or swelling,; beta 4 and / or its recurrence after surgery is avoided in a body or patient order, individual or, forgetting $ h / including humans and any other mammals, including, but Not limited to mice, gentry, guinea pigs, rabbits, miscellaneous, dogs, sheep, goats, 39 201200525 牛牛.马#子, 狒狒 and black, especially a commercially important animal or domesticated animal According to the present invention, it has been found that the peptide having the sequence identification number: 2 to 4, particularly the amino acid sequence of 3, is a hu_a2 restricted epitope peptide or candidate which can induce strong and specific An immunological reaction. Therefore, the pharmaceutical agent or composition of the present invention comprising any of the two peptides having the sequence identification number: 2 to 4', particularly the amino acid sequence of 3, is particularly suitable for administration to an individual having an antigen of HLA-A2. The same thing To a pharmaceutical agent or composition comprising a polynucleotide encoding any of the peptides (i.e., the polynucleotide of the present invention). The cancer or tumor treated by the pharmaceutical agent or composition of the present invention is not limited to and includes wherein MYBL2 ( For example, all types of cancer or tumors that are overexpressed, including 'eg, pill cancer, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. The pharmaceutical agent or composition of the present invention may include In addition to the active ingredient, there are other peptides that induce the ability of the cytotoxic lymphocytes to resist cancer-like cells, other polynucleotides encoding the other peptides, other cells expressing the other peptides, or the like. Other peptides having the ability to induce cytotoxic T lymphocytes against cancer-like cells are exemplified by cancer-specific antigens (eg, defined tumor-associated antigens), but are not limited thereto. If desired, the pharmaceutical agents of the invention or The composition may include other therapeutic substances as an active ingredient as needed, as long as the substance does not inhibit the antitumor effect of the active ingredient, the active ingredient Such as any of the peptides of the present invention. For example, the formulation may include anti-inflammatory agents, analgesics, chemotherapeutics and the like. In addition to the package 5 40 201200525 including other therapeutic substances in the agent itself, the agent of the present invention may be combined with or The other pharmaceutical agents are administered sequentially or simultaneously. The amount of the agent and the pharmaceutical agent depends on, for example, which pharmaceutical agent is used, the disease to be treated, and the manner of administration. It should be understood that, in addition to the ingredients specifically mentioned herein. In addition, the pharmaceutical agents or compositions of the present invention may include other agents generally in the art having formulation forms in question. In one embodiment of the invention, the pharmaceutical agents or compositions of the invention may be included in the manufacture. Goods and kits that contain materials useful for the pathology of the disease to be treated, such as cancer. The article of manufacture may comprise a container of any of the pharmaceutical agents or compositions of the invention having a label. Suitable containers include crucibles, vials and test tubes. The container can be formed from a variety of materials such as glass or ". The label on the container indicates that the reagent is used to treat or avoid the disease - or multiple conditions. The label may also indicate the indication of administration. In addition to the above-described containers, the kit comprising the pharmaceutical agent or composition of the present invention may further comprise, if desired, a second container which stores a pharmaceutically acceptable diluent. It may further comprise a commercial or user point of view. Other materials needed 'include other buffer solutions, diluents, buffers, needles, syringes, and package inserts with instructions for use. ° 药学 Pharmaceutical reagents or compositions can be presented in a pack or dispenser if desired 'It may comprise one or more unit dosage forms containing the active ingredient. The package may, for example, comprise a metal or _, such as a foam box (10). The pack or dispenser may be accompanied by a dosing indication. 41 201200525 (1) Pharmaceutical reagents Or the composition comprising the peptide as an active ingredient can be directly administered to the peptide of the present invention as a pharmaceutical reagent or composition, if necessary, which can be formulated by a general formulation method. In the following examples, in addition to the peptide of the present invention, It is not particularly limited as long as it is suitable to include a carrier, an excipient, and the original as a drug. An example of the above carrier is sterilized water physiological saline, phosphorus. A buffer solution and a culture fluid and the like. Further, if necessary, the pharmaceutical agent or composition may contain a stabilizer, a suspension, a preservative, a surfactant, and the like. Or the composition can be used for anti-cancer purposes. The peptide of the present invention can be prepared as a combination consisting of two or more peptides of the present invention to induce cytotoxic T lymphocytes with h W . They may be performed in the form of cocktails or may be combined with each other using standard techniques. For example, 'the peptides may be chemically linked or behave as a single fused multi-peptide sequence 歹! The winnings may be the same or different. By giving the triumph of the invention 'High-density presentation of peptides to antigen-presenting cells by HLA antigens, followed by induction of cytotoxic T lymphocytes that form a specific response to the complex between the peptide and the HLA antigen" or antigen-presenting cells (eg The dendritic cells are removed from one body and then stimulated with the peptide of the present invention to obtain antigen-presenting cells which exhibit any of the peptides of the present invention on the cell surface thereof. The cells are then administered to the individual to induce cytotoxic T lymphocytes in the individual, and thus may increase the invasion toward the tumor-associated endothelium. - A pharmaceutical agent or composition for treating and/or avoiding cancer or tumor, the package thereof The peptide of the present invention is an active ingredient, and may also comprise an adjuvant known to effectively build up the cell immunity of 42 201200525. Alternatively, the pharmaceutical agent or composition may be administered with other active ingredients or formulated. Fine particles are administered. An adjuvant refers to a compound that, when administered (or sequentially) with an immunologically active protein, enhances the anti-protein immune response. Adjuvants considered herein are included in the literature (Clin M1Cr〇b1) 〇1 Rev 1 994, 7: m 89. Examples of suitable adjuvants include aluminum phosphate, aluminum hydroxide, alum, cholera toxin, salmonella toxin, Freund's incomplete adjuvant (Inc〇mpiete) Freund*s adjuvant, 〖FA), Freund's complete adjuvant (c〇mplete

Freund’s adjuvant, CFA) 、 ISCOMatrix 、 GM-CSF 、 CpG 、 0/W乳劑與此類,但不限於此。 此外’可便利地使用於其中胜肽連結至幾個微米直徑 之小珠的微脂體(liposome)配方與細粒配方中,與於其中 脂質連結至胜肽的配方中。 在本發明另一實施例中,本發明胜肽也可以一藥學上 可接受之鹽類的形式被投予。鹽類之較佳例子包括具有驗 金屬之鹽、具金屬之鹽、具有機鹼之鹽、具有機酸之鹽與 具無機酸之鹽。如此處所使用,措辭“藥學上可接受之鹽 類”意指維持化合物生物有效性與特性及獲得自與無機酸 或驗’例如鹽酸、氫溴酸、硫酸、硝酸、填酸、甲基確酸 (methanesulfonic acid)、乙基項酸(ethanesulfonic acid)、對甲苯磺酸(p-toluenesul fonic acid)、水揚酸 (sal icy 1 ic acid)與其類似物反應的那些鹽。較佳鹽類之 例子包括具有驗金屬之鹽、具金屬之鹽、具有機鹼之鹽、 具有機酸之鹽與具無機酸之鹽。 43 201200525 在一些實施例中,本發明之藥學試劑或組合物可更包 括一成分其啟動細胞毒殺性T淋巴球。已定義脂質為可 F/叩啟動抗病毒抗原之細胞毒殺性T淋巴球的試劑。例 如,可將棕櫚酸殘基黏附至離胺酸殘基之ε-與α -胺基, 且之後連結至本發明之一胜肽。之後脂質胜肽可被直接投 予於微胞或顆粒中、併入微脂體或乳化於一佐劑中。如脂 質啟動細胞毒殺性Τ淋巴球反應之另一例子,[co / /脂 蛋白,例如三軟脂酸-S甘油半胱氨酰-絲氨酰基絲氨酸 (tripalmitoyl-S-glycerylcysternyl-seryl-serine, P3CSS)可使用來啟動細胞毒殺性τ淋巴球,當共價附加至 一合適之胜肽(參見’例如Deres et al.,Nature 1989, 342: 561-4) 〇 才又藥之方法可為口服、皮膚内、皮下、靜脈内注射或 此類,以及全身投藥或局部投藥至標的位置的鄰近區域。 可執行單··人投藥或藉由多次投藥追加。本發明之胜狀劑量 根據要治療之疾病、病患年紀、體重、投藥方法、與此類 可適合地調整,且本發明之胜肽劑量一般為〇.〇〇1 ^至 1 000 mg,例如〇. i mg至10 mg,且可於數天至數個月投 藥一次。熟悉此技藝人士可適合地選擇一合適的劑量。 (2)藥學試劑或組合才勿包含多核苦酸為活性成分 本發明之藥學試劑或組合物也可包含編碼出此處揭露 之胜肽的核酸於-可表達之形式中。此處措辭“於一可表 達之形式中”意指多核_酸,當引入—細胞叩會 被表現成-誘導抗腫瘤免疫力之多胜肽。在__代表實施例 ⑤ 44 201200525 中’感興趣之多核苷酸的核酸序列包括對於表現多核苷酸 而言必須之調控要素。可裝配多核苷酸以達到穩定插入目 標細胞之基因體(參見’例如敘述同源重組盒式載體 (cassette vector)的 Thomas KR &amp; Capecchi MR, Cell 1 987. 51 : 503-1 2 )。參見,例如 w〇l f f ei a/.,Science 1990, 247: 1465-8; U. S. Patent Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647;與 WO 98/04720) °DNA輸送技術的例子包括“裸dna” 、經促進 (bupi vacaine、聚合物、胜肽居中之)之輸送、陽離子脂 質複合物與顆粒居中之(“基因槍”)或壓力居中之傳送 (參見,例如 U.S. Patent No. 5, 922, 687 )。 本發明之胜肽也可藉由病毒或細菌載體來表現。表現 載體的例子包括減弱病毒宿主,例如牛痘或禽痘。此方法 包括使用牛痘病毒,例如為一載體以表現編碼胜肽之核苷 酸序列。藉由引入一宿主,此重組之牛痘病毒表現致免疫 胜肽且因此引起一免疫反應。於免疫步驟中為有效之牛痘 載體與方法敘述於’例如U.S. Patent No. 4, 722, 848。 另一載體為 BCG (Baci lie Calmette Guerin)。BCG 載體敘 述於 St〇ver et al.,Nature 1991,351: 456-60 中。對 於/〇療投藥或免疫有用之其他多種載體,例如腺與腺病毒 相關之載體、反轉錄病毒載體、傷寒沙門氏菌(Salm0nella ty ph 1)載體、經解毒之炭疽毒素載體與其類似為明顯的。 參見,例如 Shata ei a人,Mol Med Today 2000,6: 66-71;Freund’s adjuvant, CFA), ISCOMatrix, GM-CSF, CpG, 0/W emulsions and the like, but are not limited thereto. Further, it can be conveniently used in a liposome formulation and a fine particle formulation in which a peptide is linked to a bead of several micrometers in diameter, and in a formulation in which a lipid is linked to a peptide. In another embodiment of the invention, the peptide of the invention may also be administered in the form of a pharmaceutically acceptable salt. Preferable examples of the salt include a metal salt, a metal salt, a salt having an organic base, a salt having an organic acid, and a salt having an inorganic acid. As used herein, the phrase "pharmaceutically acceptable salts" means maintaining the biological effectiveness and properties of the compound and obtaining it from inorganic acids or assays such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, acid, and acid. (methanesulfonic acid), ethanesulfonic acid, p-toluenesul fonic acid, salic acid (sal icy 1 ic acid) and those analogs thereof. Examples of preferred salts include salts having a metal test, a salt having a metal, a salt having an organic base, a salt having an organic acid, and a salt having an inorganic acid. 43 201200525 In some embodiments, a pharmaceutical agent or composition of the invention may further comprise a component that initiates a cytotoxic T lymphocyte. Lipids have been defined as agents that can initiate cytotoxic T lymphocytes of antiviral antigens. For example, a palmitic acid residue can be attached to the epsilon- and a-amino group of an amine acid residue, and then linked to one of the peptides of the present invention. The lipid peptide can then be administered directly into the micelles or granules, incorporated into the liposomes or emulsified in an adjuvant. Another example of a lipid-inducing cytotoxic lymphocyte reaction is [co / / lipoprotein, such as tripalmitoyl-S-glycerylcysyl-seryl-serine (tripalmitoyl-S-glycerylcysternyl-seryl-serine, P3CSS) can be used to initiate cytotoxic tau lymphocytes, when covalently attached to a suitable peptide (see 'eg Deres et al., Nature 1989, 342: 561-4) , intradermal, subcutaneous, intravenous injection or the like, as well as systemic administration or topical administration to adjacent areas of the target location. It can be administered by a single person or by multiple administrations. The dosage of the present invention can be suitably adjusted depending on the disease to be treated, the age of the patient, the body weight, the administration method, and the like, and the dosage of the peptide of the present invention is generally 〇1〇〇 to 1 000 mg, for example, i. i mg to 10 mg, and can be administered once every few days to several months. Those skilled in the art will be able to suitably select a suitable dosage. (2) The pharmaceutical agent or combination does not include polynucleic acid as the active ingredient. The pharmaceutical agent or composition of the present invention may also comprise a nucleic acid in an expressible form which encodes the peptide disclosed herein. The phrase "in a form that is expressible" as used herein means a polynuclear acid which, when introduced, is expressed as a multi-peptide which induces anti-tumor immunity. The nucleic acid sequence of the polynucleotide of interest in __ represents Example 5 44 201200525 includes regulatory elements necessary for the expression of the polynucleotide. The polynucleotide can be assembled to achieve stable insertion of the target cell into the target cell (see, for example, Thomas KR &amp; Capecchi MR, Cell 1 987. 51: 503-1 2, which describes the homologous recombination cassette vector). See, for example, w〇lff ei a/., Science 1990, 247: 1465-8; US Patent Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; and WO 98/04720) Examples of DNA delivery techniques Including "naked dna", delivery (bupi vacaine, polymer, peptide centered) delivery, cationic lipid complex and particle centered ("gene gun") or pressure centered delivery (see, for example, US Patent No. 5, 922, 687). The peptide of the present invention can also be expressed by a viral or bacterial vector. Examples of performance vectors include attenuating viral hosts such as vaccinia or fowl pox. This method involves the use of a vaccinia virus, such as a vector to represent the nucleotide sequence encoding the peptide. By introducing a host, the recombinant vaccinia virus exhibits an immunogenic peptide and thus causes an immune response. The vaccinia carrier and method which are effective in the immunization step are described, for example, in U.S. Patent No. 4,722,848. Another vector is BCG (Baci lie Calmette Guerin). The BCG vector is described in St〇ver et al., Nature 1991, 351: 456-60. A variety of other vectors useful for administration or immunization, such as a gland-and adenovirus-related vector, a retroviral vector, a Salmonella tymphium (Salm0nella ty ph 1) vector, a detoxified anthrax toxin vector, and the like are apparent. See, for example, Shata ei a, Mol Med Today 2000, 6: 66-71;

Shedlock et al. , J Leukoc Biol 2000, 68: 793-806; Hipp 45 201200525 ef 3厂,/λ 2000, 14: 571-85。 輸送多核苷酸進入一個體可為直接,於其例子中,個 體直接暴露於一攜帶多核苷酸之載體,或為間接,於其例 子中,細胞首先/λ I// ire?以感興趣之多核苷酸轉形,之後 將細胞轉殖進入個體。此兩方法分別為已知,為_/yj j/·/ 與ez 7/ κσ基因治療。 基因治療之方法之大體回顧,參見Goldspiel ei a人, Clinical Pharmacy 1993, 12: 488-505; Wu and Wu, Biotherapy 1991, 3: 87-95; Tolstoshev, Ann Rev Pharmacol Toxicol 1993, 33: 573-96; Mulligan, Science 1993, 260: 926-32; Morgan &amp; Anderson, Ann Rev Biochem 1993, 62: 191-217; Trends in Biotechnology 1993,Shedlock et al., J Leukoc Biol 2000, 68: 793-806; Hipp 45 201200525 ef 3 plant, /λ 2000, 14: 571-85. The delivery of the polynucleotide into a single body can be direct, in which case the individual is directly exposed to a vector carrying the polynucleotide, or indirectly, in which case the cell is first /λ I//ire? The polynucleotide is transformed and the cells are then transferred into the individual. These two methods are known as _/yj j/·/ and ez 7/ κσ gene therapy. For a general review of methods of gene therapy, see Goldspiel ei a, Clinical Pharmacy 1993, 12: 488-505; Wu and Wu, Biotherapy 1991, 3: 87-95; Tolstoshev, Ann Rev Pharmacol Toxicol 1993, 33: 573-96 Mulligan, Science 1993, 260: 926-32; Morgan &amp; Anderson, Ann Rev Biochem 1993, 62: 191-217; Trends in Biotechnology 1993,

11 (5): 155-215)。於重組DNA技術中一般熟知的方法其也 可用於本發明,敘述於編者Ausubel ei a/,,Current Protocols in Molecular Biology, John Wi 1 ey &amp; Sons, NY, 1993; and Krieger, Gene Transfer and Expression, A11 (5): 155-215). Methods well known in the art of recombinant DNA can also be used in the present invention, described in the editor Ausubel ei a/,, Current Protocols in Molecular Biology, John Wi I ey &amp; Sons, NY, 1993; and Krieger, Gene Transfer and Expression , A

Laboratory Manual, Stockton Press, NY, 1990。 才又藥之方法可為口服、皮膚内、皮下、靜脈内注射或 此類,以及全身投藥或局部投藥至標的位置的鄰近區域提 供使用。可執行單次投藥或藉由多次投藥追加。於適合載 體中或於以編碼出本發明之胜肽的多核苷酸轉形之細胞中 的多核苷酸的劑量根據要治療之疾病、病患年紀、體重、 投藥方法、與此類可適合地調整,且本發明之胜肽劑量一 般為0. 001 mg至1〇〇0 mg,例如〇 i mg至1〇呢,且可 46 201200525 %母数天一次至备盤彻B , 母數個月一次投藥。熟悉此技藝人士可適 合地選擇一合適的劑量。 X·使用胜狀、外斗/丨、牌 .Laboratory Manual, Stockton Press, NY, 1990. The method of medicinal administration can be administered orally, intradermally, subcutaneously, intravenously, or the like, as well as in the vicinity of systemic administration or topical administration to a targeted location. A single administration can be performed or by multiple administrations. The dosage of the polynucleotide in a suitable vector or in a cell transformed with a polynucleotide encoding the peptide of the present invention may suitably be according to the disease to be treated, the age of the patient, the body weight, the method of administration, and the like. Adjusted, and the dosage of the peptide of the present invention is generally from 0.001 mg to 1 〇〇 0 mg, for example, 〇i mg to 1 〇, and can be 46 201200525 % mother days once to prepare for B, mother months Give it once. Those skilled in the art will be able to suitably select a suitable dosage. X·Use wins, outer buckets/丨, cards.

广吐j、體、抗原呈現細胞與細胞毒殺性T 淋巴球的方法 可使用本發明之胜狀與多核苦酸來製備或誘導抗原呈 現細胞與細胞毒殺性τ淋巴球。也可使用本發明之外吐小 與抗原呈現細胞來誘導細胞毒殺性T淋巴球。胜狀、多 苷文外吐小體與抗原呈現細胞可與任何其他化合物结 口使用,只要化合物不抑制其細胞毒殺性T淋巴球誘發能 力因此,任何上述之本發明藥學試劑或組合物可用來誘 導’田胞毒殺性τ淋巴球’且除此之外,包括胜肽與多核苷 复的那二也可用來誘導抗原呈現細胞,如於下所討論。 (1)誘導抗原呈現細胞的方法 本發明提供使用本發明之胜肽或多核苷酸來誘導具有 高細胞毒殺性τ淋巴球誘發能力之抗原呈現細胞的方法。 本發明之方法包括M &quot;叩或h 將 抗原呈現細胞與本發明胜肽接觸的步驟。例如,h叩 將抗原呈現細胞與胜肽接觸的方法可包括步驟: a :自一個體收集抗原呈現細胞;以及 b:將步驟a之抗原呈現細胞與胜肽接觸。 抗原呈現細胞並不限於特定種類之細胞,且包括樹突 細胞、蘭格罕細胞(Langerhans ceU)、巨嗜細胞、B細胞 與活化之τ細胞,已知其表現蛋白質(pr〇teinace〇us)抗原 於其細胞表面以被淋巴球所辨認。可較佳使用樹突細胞, 47 201200525 由於匕們於抗原呈現細胞中之最強的細胞毒殺性τ淋巴球 誘發能力。本發明任何胜肽可被使用為步驟b之胜肽以它 們本身或與本發明其他胜肽一起。 或者’可投予本發明胜肽至一個體以使抗原呈現細胞 Η FC?與胜肽接觸。因此可於個體之體内誘導具有高細 胞毒殺性Τ淋巴球誘發能力之抗原呈現細胞。因此,本發 明也考慮投予本發明胜肽至一個體以/万叩誘導抗原呈 現細胞的方法。也可能投予編碼出本發明胜肽之多核苷酸 至一個體於一可表達之形式中,以便本發明胜肽被表現且 』刀F/ F0與抗原呈現細胞接觸,以因此於個體之體内誘導 具有间細胞毒殺性Τ淋巴球誘發能力之抗原呈現細胞。因 此’本發明也考慮投予本發明多核苷酸至一個體以“ h叩 誘導抗原呈現細胞的方法。措辭“可表達之形式,,被定義 於上述段落‘IX.藥學試劑或組合物藥學試劑包含 多核苷酸為活性成分”中。 此外,本發明可包括將本發明多核苷酸引入一抗原呈 現細胞以便誘導具有細胞毒殺性T淋巴球誘發能力之抗原 呈現細胞。例如,方法可包括步驟: a :自一個體收集抗原呈現細胞;以及 b :將編碼出本發明胜肽之一多核苷酸引入。 可如引述^又落VI _抗原呈現細胞,,中所述來執行步 驟b。 (2)誘導細胞毒殺性τ淋巴球的方法 本發明也提供使用本發明胜肽、多核苷酸、外吐小體 48 201200525 或抗原呈現細胞來誘導細胞毒殺性τ淋巴球的方法。 本發明也提供使用編碼出一多胜肽之多核苷酸來誘導 細胞毒殺性τ淋巴球的方法,此多胜肽具形成一 τ細胞受 體次單位的能力,而此τ細胞受體次單位辨認一本發明胜 狀與HLA抗原之複合物。較佳為,誘導細胞毒殺性τ淋巴 球的方法包括至少一步驟擇自由下列之中: a) 將一 CD8陽性Τ細胞與一抗原呈現細胞及/或一外 吐小體接觸,該抗原呈現細胞及/或該外吐小體表現一 HLA 抗原與本發明胜肽之複合物於其表面上,以及 b) 將一多核苷酸引入一 CD8陽性T細胞,其中該多核 苷酸編碼出一多胜肽,該多胜肽具形成一 τ細胞受體次單 位的旎力,而該τ細胞受體次單位辨認一本發明胜肽與HLA 抗原之複合物。 當本發明之胜肽、多核苷酸、抗原呈現細胞或外吐小 體被投予至一個體時,於個體體内誘導細胞毒殺性τ淋巴 球,且以癌細胞為目標之免疫反應的強度增強。因此,本 發明也考慮一方法,其包括將本發明之胜肽、多核苷酸、 抗原呈現細胞或外吐小體投予至一個體以誘導細胞毒殺性 T淋巴球的步驟。 或者,藉由它們的ex h 使用,也可誘導細胞毒殺 性τ淋巴球。於此類例子中’在誘導細胞毒殺性τ淋巴球 後,經活化之細胞毒殺性τ淋巴球可返回至個體。例如, 誘導細胞毒殺性τ淋巴球之本發明方法可包括步驟: a):自一個體收集抗原呈現細胞; 49 201200525 b):將步驟a)之抗原呈現細胞與胜肽接觸;以及 將步驟b之抗原呈現細胞與CD8陽性細胞共培養。 於上述步驟c中要與⑽陽性細胞共培養之抗原呈現 細胞也可藉由將一包括本發明多核苦酸之基因轉移進入抗 原呈現細胞,如於前述段落“ VI.抗原呈現細胞,,中所述 j製備;然而,本發明並不限於此’且包括任何有效表現 - HLA抗原與本發明胜肽之複合物於其表面上的抗原呈現 細胞。 代替此種抗原呈現細胞,也可使用呈現一心抗原與 本發明胜肽之複合物於其表面上的外吐小體。換句話說, 本發明也考慮-方法,其令將呈現一 HU抗原與本發明胜 肽之複合物於其表面上的外吐小體與⑽陽性細胞共培 養。此種外吐小體可藉由前述於段落“V.夕卜土小體,,中之 方法來製備。 藉由將包括編碼出與本發明一胜肽結合之T 胞又體-人單兀的多核苷酸的基因引入⑽陽性細胞可誘 導、·田胞毒殺性T淋巴球。如於前述段《“VIJ. τ細胞受 體(TCR)”中所述可執行此轉導。 此卜本發明也提供製造一誘導細胞毒殺性τ淋巴球 之藥學試劑或組合物的方法或製程,其中該方法包括將本 發月之胜肽與藥學上接受之載體一起混合或配製的步驟。 (3)誘導免疫反應的方法 又’本發明提供誘導抗峨2相關疾病之免疫反應的 適。的疾病包括癌症,其例子包括,但不限於睪丸 50 201200525 小細胞肺癌與食 腫瘤、胰臟癌、膀胱癌、非小細胞肺癌 道癌。 本發明方法可包括投予含任何本發明胜肽或編碼出其 夕核苷i的4劑或組合物的步驟。本發明方法也考慮投 予表現任何本發明胜肽之外吐小體或抗原呈現細胞。二節 參見“IX.藥學試劑或組合物”之項目,特別是敘述本發 明試劑或組合物為疫苗之用途的部分。此外,可被使用於 本發明誘導免疫反應之方法的外吐小體與抗原呈現細胞, 被詳細描述在前之“v.夕卜吐小體’’、“νι· &amp;原呈現細 胞與X.使用胜狀、外吐小體、抗原呈現細胞與細胞毒 殺性Τ淋巴球的方法”之⑴與⑺的項目。 本發明更提供在-個體中誘導抗癌;症,例如畢丸腫 瘤、胰臟癌、膀胱癌、非小細胞肺癌、小細胞肺癌與食道 癌之免疫反應的方法。為此目的,本發明包括製造一用於 誘導免疫反應之藥學試劑、物質或組合物的方法,其中該 方法包括將本發明之胜肽或多核苷酸與藥學上接受之載體 一起混合或配製的步驟。或者,用於誘導免疫反應的方法 包括配製並投予本發明一疫苗的步驟,此類疫苗包括: (a) —或多個本發明抗原決定位胜肽,或一其免疫活 性片段; (b) —或多個編碼出(a)之抗原決定位胜肽或免疫活 性片段之多核苷酸; (c) 一或多個之本發明經分離之細胞毒殺性τ淋巴 球;或 51 201200525Method for stimulating j, body, antigen-presenting cells and cytotoxic T lymphocytes The use of the present invention and polynucleic acid can be used to prepare or induce antigen-presenting cells and cytotoxic T lymphocytes. The extracorporeal and antigen-presenting cells of the present invention can also be used to induce cytotoxic T lymphocytes. The phenotype, the polyglucoside and the antigen presenting cells can be used in combination with any other compound, as long as the compound does not inhibit its cytotoxic T lymphocyte evoking ability, any of the above-mentioned pharmaceutical agents or compositions of the present invention can be used Induction of 'field cytotoxic tau lymphocytes' and in addition, the second including the peptide and the polynucleoside complex can also be used to induce antigen-presenting cells, as discussed below. (1) Method for inducing antigen-presenting cells The present invention provides a method of inducing antigen-presenting cells having high cytotoxic tau lymphocyte-inducing ability using the peptide or polynucleotide of the present invention. The method of the present invention comprises the step of contacting the antigen presenting cells with the peptide of the present invention by M &quot;叩 or h. For example, the method of contacting the antigen presenting cells with the peptide may comprise the steps of: a: collecting antigen presenting cells from one body; and b: contacting the antigen presenting cells of step a with the peptide. The antigen presenting cells are not limited to a specific kind of cells, and include dendritic cells, Langerhans cells (cegerhans ceU), macrophages, B cells, and activated tau cells, which are known to express proteins (pr〇teinace〇us). The antigen is recognized by the lymphocytes on its cell surface. Dendritic cells can be preferably used, 47 201200525 because of their strongest cytotoxic thymocyte-inducing ability in antigen-presenting cells. Any of the peptides of the invention can be used as the peptide of step b either by themselves or with other peptides of the invention. Alternatively, the peptide of the present invention can be administered to a body such that the antigen exhibits cell Η FC? in contact with the peptide. Therefore, an antigen-presenting cell having a high cytotoxic lymphocyte-inducing ability can be induced in an individual. Accordingly, the present invention also contemplates a method of administering a peptide of the present invention to a body to induce antigen-presenting cells. It is also possible to administer a polynucleotide encoding the peptide of the present invention to an expressible form such that the peptide of the present invention is expressed and the knife F/F0 is brought into contact with the antigen presenting cells, thereby An antigen-presenting cell having an ability to induce cytotoxic lymphocyte inducing cells is induced. Thus, the present invention also contemplates the administration of a polynucleotide of the present invention to a single body to "h叩 induce antigen-presenting cells. The expression "expressible form," is defined in the above paragraph 'IX. Pharmaceutical reagent or composition pharmaceutical reagent Further, the present invention may include introducing the polynucleotide of the present invention into an antigen-presenting cell to induce an antigen-presenting cell having a cytotoxic T lymphocyte-inducing ability. For example, the method may include the steps of: a: collecting antigen-presenting cells from one body; and b: introducing a polynucleotide encoding one of the peptides of the present invention. Step b can be performed as described in the reference to the VI-antigen-presenting cells. 2) Method for Inducing Cytotoxic Thirteen Lymphocytes The present invention also provides a method for inducing cytotoxic tau lymphocytes using the peptide, polynucleotide, exosome 48 201200525 or antigen presenting cells of the present invention. The present invention also provides A method for inducing a cytotoxic tau lymphocyte using a polynucleotide encoding a multi-peptide, the ability of the multi-peptide to form a tau cell receptor subunit And the tau cell receptor subunit recognizes a complex of the invention and the HLA antigen. Preferably, the method for inducing the cytotoxic tau lymphocyte comprises at least one step of selecting the following: a) positing a CD8 The sputum cell is contacted with an antigen presenting cell and/or an exosome, the antigen presenting the cell and/or the exosome exhibiting a complex of an HLA antigen and a peptide of the present invention on its surface, and b) A polynucleotide is introduced into a CD8-positive T cell, wherein the polynucleotide encodes a multi-peptide that has a force to form a tau cell receptor subunit, and the tau cell receptor subunit identification A complex of a peptide of the invention and an HLA antigen. When the peptide, polynucleotide, antigen-presenting cell or exosome of the present invention is administered to a body, the cell toxic lymphocytes are induced in the body. And the intensity of the immune response targeting cancer cells is enhanced. Therefore, the present invention also contemplates a method comprising administering a peptide, a polynucleotide, an antigen presenting cell or an exosome of the present invention to a body. Induced cytotoxic T-leaching The step of the ball. Or, by their use of ex h, it can also induce cytotoxic thymocytes. In such cases, 'inactivated cytotoxic lymphocytes after inducing cytotoxic T lymphocytes Returning to the individual. For example, the method of the invention for inducing cytotoxic tau lymphocytes can include the steps of: a) collecting antigen presenting cells from one body; 49 201200525 b): bringing the antigen of step a) to the cell and contacting the peptide And co-culturing the antigen-presenting cells of step b with CD8-positive cells. The antigen-presenting cells to be co-cultured with (10) positive cells in the above step c can also be obtained by transferring a gene including the polynucleic acid of the present invention into an antigen. The cell, as prepared in the aforementioned paragraph "VI. Antigen presenting cells, described in j; however, the invention is not limited thereto" and includes any effective expression - a complex of the HLA antigen and the peptide of the present invention on its surface The antigen presents the cells. Instead of such an antigen presenting cell, an exosome expressing a complex of a single-heart antigen and a peptide of the present invention on its surface may also be used. In other words, the present invention also contemplates a method in which an exosome having a complex of a HU antigen and a peptide of the present invention on its surface is co-cultured with (10) positive cells. Such exosomes can be prepared by the method described in the above paragraph "V. U.S. s., by including the T-cell-human-single that encodes a peptide of the present invention. The gene of the polynucleotide is introduced into (10) positive cells to induce, cytotoxic T lymphocytes. This transduction can be performed as described in the aforementioned paragraph ""VIJ. τ cell receptor (TCR)". The invention also provides a method or process for the manufacture of a pharmaceutical agent or composition for inducing a cytotoxic tau lymphocyte, wherein the method comprises the step of mixing or formulating the peptide of the present month with a pharmaceutically acceptable carrier. Method for Inducing Immune Response Further, the present invention provides a suitable disease for inducing an immune response against a disease associated with 峨2, including cancer, and examples thereof include, but are not limited to, testicular 50 201200525 small cell lung cancer and oncology, pancreatic cancer, bladder cancer Non-small cell lung cancer tract cancer. The method of the invention may comprise the step of administering 4 doses or compositions comprising any of the peptides of the invention or encoding ceramide nucleoside i. The method of the invention also contemplates administration of any of the inventions Peptide outside the peptide Or antigen presenting cells. Section II refers to the item "IX. Pharmaceutical Reagents or Compositions", particularly the part in which the reagent or composition of the present invention is used for a vaccine. Further, it can be used in the method of inducing an immune response of the present invention. The exocytosis and antigen-presenting cells are described in detail in the previous "v. Xi Bu Tu Body", "νι· &amp; original cells and X. use wins, exosome, antigen presenting cells and The method of cytotoxic sputum lymphocytes" (1) and (7). The present invention further provides a method of inducing an anti-cancer disease in an individual, such as a pill tumor, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. To this end, the invention includes a method of making a pharmaceutical agent, substance or composition for inducing an immune response, wherein the method comprises mixing or formulating a peptide or polynucleotide of the invention together with a pharmaceutically acceptable carrier step. Alternatively, the method for inducing an immune response comprises the step of formulating and administering a vaccine of the invention, the vaccine comprising: (a) - or more antigenic epitope peptides of the invention, or an immunologically active fragment thereof; - or a plurality of polynucleotides encoding the epitope of the (a) peptide or immunologically active fragment; (c) one or more of the isolated cell toxic lymphocytes of the invention; or 51 201200525

Cd) —或多個之本發明經分離之抗原呈現細胞。 在本發明内容中’以這些活性成份可治療過度表現 MYBL2之癌症。此類癌症的例子包括,但不限於睪丸腫瘤、 胰臟癌、膀胱癌、非小細胞肺癌、小細胞肺癌與食道癌。 因此,在包括活性成分之疫苗或藥學組合物的投予前,其 較佳為確認與相同器官之正常組織相較,MYBL2之表現程 度於要被治療之癌細胞或組織中是否被提高。因此,在— 實施例中,本發明提供治療(過度)表現MYBL2之癌症的 方法’其方法可包括步驟: i)測疋獲得自具有癌症要治療之個體的癌細胞或组 織中的MYBL2表現程度; … 1 i )與正常控制組比較MYBL2表現程度;以及 ui)投予擇自由上述(3)至((1)所組成之群組的至少 一成份至與正常控制組相較具有過度表現mybl2之癌症的 個體。 或者,本發明也可提供包含擇 組的至少—成份的疫苗或藥學組合物,其用於投予至具 過度表現隨2之癌症的個體中。換句話說,本發明更 供鑑定要被以本發明贿2多胜肽治療之個體的方法, 類方法包括測;t來自個體之癌細胞或組織中的峨2表 程度的步驟,其中與基因之正常控制程度相較,此程度」 加指出個體具有可以本發明嶋多胜肽治療之癌症。 發明之治療癌症的方法於以下更詳細敘述。 要藉由本發明治療之個^^击 縻之個體較佳為一哺乳類動物。示摩 52 201200525 之哺乳類動物包括,但不限於,例如, 類動物、小鼠、大鼠、狗、描、馬與牛。員非人類靈長 根據本發明,測定獲得自一個體之 的MYBL2表現程度。使用本技術 ,·,I或組織中 酸)產物程度測定表現程度。例:域藉已::可於轉錄(核 相田雜合方法「办丨l 北方雜合)使用探針可將訂犯的量。可於—, 陣列上執行㈣。陣列之使用較佳為用於偵測二 度。利用舰2的序列資訊,熟悉此技藝人士可製 備此種探針。例如,瞧2的峨可被使用為探針。若需 要’可以適合之標誌來標誌探針,例如染劑、螢光物質與 同位素’且基因的表現程度可被谓測為雜合標誌的強度:、 可將任何源自個體之細胞或組織用於mybl2表現之測 定’只要其包括MYBL2之目標轉錄或轉譯產物。適合樣本 的例子包括,但不限於身體組織或液體、例如血液、唾液 與尿液。較佳為,源自個體之細胞或組織包含一細胞族群, 其包括一上皮細胞,更佳為一癌症上皮細胞或一來自被懷 疑癌化之組織的上皮細胞。此外,若需要,細胞可自所獲 得之身體組織或液體被純化,且之後使用為源自個體之樣 本。藉由擴大偵測方法(amplification-base detectin method)(例如,RT-PCR)使用引子可將MYBL2的轉錄產物 進行定量。根據基因之可獲得序列資訊可製備此種引子。 特別是,用於本方法之探針或引子於嚴厲 (stringent)、適度嚴厲、低嚴厲條件下雜合至MYBL2的 mRNA。如此處使用,措辭“嚴厲(雜合)條件”意指在此 53 201200525 在條件下探針或引子會雜合至其目標序列,而不是其他序 列°嚴厲條件為序列依賴(seqUence_dependent),且在不 同環境下會不同。比起較短之序列,於較高溫度下觀察到 較長序列之特定雜合。一般而言,在一定義之離子強度與 pH下所選擇之嚴格條件的溫度為低於一特定序列之熔點 (Tm)約5°C。Tm為溫度(在一定義之離子強度與pH與核酸 濃度下)’於其下在平衡下5〇%之與互補目標序列的探針 雜合至目標序列。由於目標序列通常存在過量,所以於Tm, 在平衡下50%之探針被佔據。一般而言’嚴苛條件為於其 中鹽濃度低於1.0 Μ鈉離子,一般約〇.〇1至1.〇 μ鈉離 子(或其他鹽)於pH 7. 0至8. 3,且對於短探針或引子(例 如’ 1 0至5 0個核苷酸)而言溫度為至少約3 〇。〇,對於較 長探針或引子而&amp;溫度為至少約6 〇 °c。也可以添加去穩定 °式 wj (destabi 1 izing agent),例如甲醯胺(formamide)來 達到嚴苛條件。 探針或引子可為特定大小。大小的範圍始於至少丨〇個 核苷酸、至少12個核苷酸、至少15個核苷酸、至少2〇個 核苷酸、至少25個核苷酸、至少3〇個核苷酸,且探針與 引子延伸大小始於5-1 〇個核苷酸、1 〇_丨5個核苷酸、1卜2〇 個核普酸、20-25個核苷酸與25-30個核苦酸。 或者’為了本發明之診斷可偵測轉譯產物。例如,可 測定MYBL2蛋白質(序列辨識號:6)之量。測定作為轉錄 產物之蛋白質的量的方法包括免疫分析方法,其使用—抗 體專一辨認此蛋白質。抗體可為單株或多株。此外,抗體 54 201200525 之任何片段或修飾(例如嵌合型抗體(chimeric antibody)、scFv、Fab、F(ab,)2、Fv 等)可被用來偵測, 只要片段或經修飾之抗體維持對MYBL2蛋白質的結合能 力。用於蛋白質偵測之這些種類的抗體的製備方法為本技 術領域所熟知,且任何方法可被使用於本發明中以製備此 種抗體與其等同物(equ i va 1 ent)。 如根據MYBL2基因轉譯產物偵測MYBL2基因之表現程 度的另一方法,使用抗MYBL2蛋白質之抗體經由免疫組織 化學(immunohistochemical)分析可測量到.染色強度。即, 於此測里中,強的染色指出增加之蛋白質存在/程度,且同 時MYBL2基因之高表現程度。 可確認於癌症細胞中’例如mYBL2基因之目標基因的 表現权度為被提升’若其相較於目標基因之控#組程度(例 如’於正常細胞中的程度)增加,例如i 、⑽、或 %,或增加大於hl倍、大於1.5倍、大於2.0倍、大於 5. 〇倍、大於1〇. 〇倍或更多。 藉由使用先刖自一個體/其疾病階段(癌的或非癌的) 為已知的個體收隼並德左&amp; 果1^存的樣本,控制組之程度可與癌細 胞同時測定。此外,獲得白 于自具有癌症要被治療之一器官的 非癌區域的正常細胎祐蚀田劣 ,甘 、破使用為正常控制組。或者,根據獲 自刀析先則測疋之來自其疾病程度已知之個體之樣本中 之隨2基因的表現程度的結果,藉由統計方法,可測定 控制組之程度。此外,护 Γ 徑制組程度可為來自自先前測試細 胞之表現輪廓的資料庫。廿β 叶車 並且,根據本發明一方面,於一 55 201200525 生物樣本中之MYBL2基因的表現程度,可與多個控制组程 度比較,其控制組程度被測定自多個參考樣本。較佳為使 用一控制組程度測定自一參考樣本,其來自一組織形式相 似於源自個體生物樣本之組織形式。此外,較佳為使用且 有已知疾病階段之群組中的MYBL2基因的表現程度的標準 值(standard value)。標準值可獲得自本技術領域任何已 知的方法。例如,平均值仏2標準差或平均值+/_3標準 差’可被使用為標準值。 本發明之内容中,測定自P 4盔 疋目已知為非癌症之生物樣本的 控制組程度被意指為- “正常控制組程度” m 若控制組程度測定自一癌的生物組織,其意指為一 “癌的 控制組程度,,。可將介於樣本表現程度與控制組程度間的 不同標準化至控制核酸的表現程度,例如管家基因 (ousekeeping gene),根據細胞之癌症與非癌程度已知其 又並…、不同。示範之控制基因包括,但不限於々肌 動蛋白(beta actin)、甘油駿_3_磷酸去氫酶Cd) - or a plurality of isolated antigens of the invention present cells. In the context of the present invention, cancers that overexpress MYBL2 can be treated with these active ingredients. Examples of such cancers include, but are not limited to, testicular tumors, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. Therefore, prior to administration of the vaccine or pharmaceutical composition comprising the active ingredient, it is preferred to confirm whether the expression of MYBL2 is increased in the cancer cells or tissues to be treated compared to the normal tissues of the same organ. Thus, in an embodiment, the invention provides a method of treating (over-expressing) a cancer of MYBL2, the method comprising the steps of: i) measuring MYBL2 expression obtained from cancer cells or tissues having an individual to be treated by the cancer Degree; ... 1 i) comparing the degree of MYBL2 performance with the normal control group; and ui) selecting at least one component of the group consisting of (3) to (1) above to have excessive performance compared with the normal control group The individual of the cancer of mybl2. Alternatively, the invention may also provide a vaccine or pharmaceutical composition comprising at least one component selected for administration to an individual having an over-expressed cancer. In other words, the invention Further, it is a method for identifying an individual to be treated by the present invention, and the method includes the step of measuring the degree of 峨2 in the cancer cells or tissues of the individual, wherein the degree of normal control of the gene is compared , to the extent that the individual has a cancer that can be treated by the multi-peptide of the present invention. The method of treating cancer of the present invention is described in more detail below. The individual to be treated by the present invention is more It is a mammal. The mammals of Momo 52 201200525 include, but are not limited to, for example, animals, mice, rats, dogs, horses, horses and cattle. Non-human primates are determined according to the present invention. The degree of MYBL2 expression of an individual. The degree of performance was determined using the technique, the degree of product in the I, or the acid in the tissue. Example: domain borrowing:: can use the probe to transcribe (nuclear phase hybrid method "handling l northern hybrid") can use the probe to execute the amount of the offense. It can be executed on the array (4). The use of the array is preferably used. For detection of the second degree. Using the sequence information of the ship 2, those skilled in the art can prepare such a probe. For example, the 瞧 2 峨 can be used as a probe. If a suitable marker can be used to mark the probe, for example Dyes, fluorescent substances and isotopes' and the degree of expression of the gene can be said to be the intensity of the heterozygous marker: any cell or tissue derived from the individual can be used for the determination of mybl2 expression as long as it includes the target transcription of MYBL2 Or translation products. Examples of suitable samples include, but are not limited to, body tissues or liquids, such as blood, saliva, and urine. Preferably, the cells or tissues derived from the individual comprise a cell population comprising an epithelial cell, preferably It is a cancer epithelial cell or an epithelial cell from a tissue suspected of being cancerized. Further, if necessary, the cell can be purified from the obtained body tissue or liquid, and then used as a sample derived from the individual. An amplification-base detectin method (eg, RT-PCR) can be used to quantify the transcript of MYBL2 using primers. Such primers can be prepared based on the available sequence information of the gene. In particular, for use in the method. Probes or primers are heterozygous to MYBL2 mRNA under stringent, moderately stringent, low stringency conditions. As used herein, the phrase "stringent (heterozygous) conditions" means that at this time, 53 201200525 probes or primers under conditions Will be heterozygous to its target sequence, rather than other sequences. Severe conditions are sequence dependent (seqUence_dependent) and will be different under different circumstances. Compared to shorter sequences, specific heterozygous sequences of longer sequences are observed at higher temperatures. In general, the temperature of a stringent condition selected at a defined ionic strength and pH is about 5 ° C below the melting point (Tm) of a particular sequence. Tm is the temperature (in a defined ionic strength versus pH and nucleic acid concentration) Lower) '5% of the probes under the equilibrium with the complementary target sequence are hybridized to the target sequence. Since the target sequence is usually present in excess, at Tm, under equilibrium 50 % of the probe is occupied. Generally speaking, the 'stringent condition is that the salt concentration is less than 1.0 Μ sodium ion, generally about 〇. 〇1 to 1. 〇μ sodium ion (or other salt) at pH 7.8 to 8. 3, and for short probes or primers (eg '10 to 50 nucleotides) the temperature is at least about 3 〇. 〇, for longer probes or primers &amp; temperature is at least about 6 〇°c. It is also possible to add destabi 1 izing agent, such as formamide, to meet stringent conditions. Probes or primers can be of a specific size. The range of sizes starts at least one. Nucleotide, at least 12 nucleotides, at least 15 nucleotides, at least 2 nucleotides, at least 25 nucleotides, at least 3 nucleotides, and the probe and primer extensions begin at 5-1 nucleotides, 1 〇 _ 5 nucleotides, 1 〇 2 nucleotides, 20-25 nucleotides and 25-30 nucleotides. Alternatively, the translation product can be detected for the diagnosis of the present invention. For example, the amount of MYBL2 protein (SEQ ID NO: 6) can be determined. A method for determining the amount of a protein as a transcription product includes an immunoassay method which specifically recognizes the protein using an antibody. The antibody may be single or multiple plants. In addition, any fragment or modification of antibody 54 201200525 (eg, chimeric antibody, scFv, Fab, F(ab,) 2, Fv, etc.) can be used to detect, as long as the fragment or modified antibody is maintained Binding ability to MYBL2 protein. Methods for the preparation of these classes of antibodies for protein detection are well known in the art, and any method can be used in the present invention to prepare such antibodies and their equivalents (equ i va ent). For another method for detecting the degree of expression of the MYBL2 gene based on the MYBL2 gene translation product, the staining intensity can be measured by immunohistochemical analysis using an antibody against the MYBL2 protein. That is, in this assay, strong staining indicates the presence/degree of increased protein and the high degree of expression of the MYBL2 gene. It can be confirmed that the expression weight of the target gene such as the mYBL2 gene is increased in cancer cells if it is increased compared to the degree of control group of the target gene (for example, 'degree in normal cells), for example, i, (10), Or %, or increase greater than hl times, greater than 1.5 times, greater than 2.0 times, greater than 5. 〇 times, greater than 1 〇. 〇 times or more. The degree of the control group can be determined simultaneously with the cancer cells by using a sample that is previously known from the individual/the disease stage (cancerous or non-cancerous) and the left and the left. In addition, the normal fine-feathers that were obtained from the non-cancer areas of the organs to be treated with cancer were inferior, and were used as the normal control group. Alternatively, the degree of the control group can be determined by statistical methods based on the results of the degree of expression of the 2 genes in the sample from the individual whose disease degree is known by the knife analysis. In addition, the degree of tamper formation can be a database of performance profiles from previous test cells. Further, according to one aspect of the present invention, the degree of expression of the MYBL2 gene in a biological sample of a 55 201200525 can be compared with a plurality of control group degrees, and the degree of the control group is determined from a plurality of reference samples. Preferably, a control panel is used to determine a reference sample from a tissue form that resembles a tissue form derived from an individual biological sample. Furthermore, it is preferred to use a standard value of the degree of expression of the MYBL2 gene in a group of known disease stages. Standard values are available from any method known in the art. For example, the average value 仏 2 standard deviation or the average value +/_3 standard deviation ' can be used as the standard value. In the context of the present invention, the degree of control of a biological sample known to be non-cancer from P 4 is considered to be - "normal control group degree" m if the control group is determined from a biological tissue of a cancer, Means a degree of cancer control, which can be normalized to the degree of expression of the control group to control the degree of expression of the nucleic acid, such as the ousekeeping gene, cancer and non-cancer according to the cell The degree is known to be... and different. Exemplary control genes include, but are not limited to, beta actin, glycerol _3_phosphate dehydrogenase

Cglyceraldehyde-3-phosphate dehydrogenase)^ ^ ^ ^ 白P卜 當與正常控制組程度相較MYBL2基因的表現程度被增 或相A /等同於癌控制組程度,可診斷個體為具有癌症要 被治療。 本發明也提供⑴診斷或確認是否一個體被懷疑具有 要被治療之癌症,刀/ 及/次(11)選擇要癌症治療之個體的方 法,其方法包括步驟·· 201200525 a) 測定在癌症鈿胞或組織中,MYBL2的表現程度癌 症細胞或組織獲得自被懷疑具有要被治療之癌症的個體; b) 與正常控制組比較MYBL2之表現程度; 〇若醜2之表現程度與正常控制組程度相較為被 增加’則診斷個體為具有要被治療之癌症;以及 d)若個體於㈣c)中被料為具有要被治療之癌 症則選擇或確認要癌症治療之個體。 或者’此種方法可包括步驟: a) 測定在癌症細胞或組織中,MYBL2的表現程度,癌 症細胞或組織獲得自被懷疑具有要被治療之癌症的個體; b) 與癌症控制組比較MYBL2之表現程度; c) 若MYBL2之表現程度相似或等於癌症控制組程 度,則診斷個體為具有要被治療之癌症;以及 d) 若個體於步驟c)中被診斷為具有要被治療之癌 症’則選擇或確認要癌症治療之個體。 本發明也k供一套組以測定一個體遭受可被以本發明 MYBL2多胜肽治療之癌症,其也在評估及/或監控癌症免疫 〜療的功效中為有用的。較佳為,癌症包括,但不限於睪 丸腫瘤、胰臟癌、膀胱癌、非小細胞肺癌、小細胞肺癌與 食道癌。更特別的是,套組較佳包括至少一用以偵測來自 個體癌細胞中之MYBL2基因的表現程度的試劑,其試劑可 被擇自群組: (a) —試劑用以偵測MYBL2基因的mRNA ; (b) —試劑用以偵測MYBL2蛋白質;以及 57 201200525 (C) 一試劑用以偵測MYBL2蛋白質的生物活性。 適合用以偵測MYBL2基因之mRNA之試劑的例子可包括 核酸其專一結合或辨認MYBL2 mRNA,例如,具有對於心此2 mRNA之一部分互補的序列的募核苷酸。這些種類之寡核苷 酸以專-於瞧2 mRNA之引子與探針為例子。根據本技術 領域所熟知的方法可製備這些種類之募核苷酸。若需要, 用以偵測MYBL2 mRNA之試劑可被固定於固體基質(贴计 上。此外,大於一個之用以偵測MYBL2 mRNA的試劑可被包 含於套組中。 另一方面,適合用以偵測MYBL2蛋白質之試劑的例子 包括對於MYBL2蛋白質的抗體。抗體可為單株或多株。此 外,抗體之任何片段或修飾(例如嵌合型抗體(chhehc antibody)、ScFv、Fab、F(ab,)2、Fv 等)可被用來作為 試劑,只要片段或經修飾之抗體維持對MYBL2蛋白質的結 合能力。用於蛋白質偵測之這些種類的抗體的製備方法為 本技術領域所熟知,且任何方法可被使用於本發明中以製 備此種抗體與其等同物(equivalent)e另外,可以訊號產 生分子經由直接連接或一間接標誌技術來將抗體進行標 誌。標誌與標誌抗體之方法與偵測抗體對其目標的結合為 本技術領域所熟知,且任何標誌與方法可被使用於本發 明。另外,大於一個之用於偵測MYBL2蛋白質的試劑可被 包括於套組中。 套組可包含多於一個之前述試劑。例如,獲得自沒有 癌症或遭受癌症之個體的組織樣本可作為有用的控制組試 ⑤ 58 201200525 劑。本發明之套組可更包括商業或 韦及使用者角度所需之其他 材枓,包括緩衝溶液、稀釋液清 愿15 /主射針、注射器與 具有使用之操作指南的包裝插入物(例如,f面、 —等)。這些試劑或此類可保持於一具有標諸之容:。 適合之容器包括瓶子、小玻璃瓶(〜υ與試驗試管。容器 可形成自多樣化之材料,例如玻璃或塑膠。 在本發明—貫施例中’當試劑為抗㈣心驗之探針 時,試劑可被固定於一固體基質上,例如一多孔條(― …⑻以形成至少-㈣位。多孔條之測量或制區可包 括複數個位置’各含有一核酸(探針)。一測試條也可含 有負及/或正控制組的位置。或者,控制組之位置可位於與 測試條分離之一條。滿兩&amp; — 視而要而疋’不同之偵測位可包含不 同量之經固定之核酸’即一較高量於第叫貞測位中且一較 低含量於隨後之位置令。藉由測試樣本的加入,顯示可偵 ^號之一些位置提供一於樣本中MYBL2mRNA存在之量的 定量指示1測位可被設置於任何適合之可偵測形狀且一 般為在橫跨—測試條之寬度的條狀物或點的形狀中。 本發明之套組可更包括一正控制組樣本或MYBL2標準 樣本。藉由收集舰2陽性之樣本且之後分析它們的醜2 程度可製備本發明之正控制組樣本。或者,可將經純化之 MYBL2蛋白質或多核苦酸加至不表現隨2之細胞以形成 正樣本(positive sampie)或MYBL2標準樣本。於本發明 中a純化之MYBL2可為重組蛋白質。正控制組樣本之 _2程度為,例如,大於臨界值(cut〇ff vaiue)。 59 201200525 在一實施例中,本發明更提供一診斷套組,包括一蛋 白質或其一部份蛋白質,具有專一辨認本發明抗體或其片 段之能力。 本發明之蛋白質之部分胜肽的例子包括多胜肽,其係 由在本發明蛋白質之胺基酸序列中之至少8個,較佳15 個、更佳20個連續胺基酸所組成。使用本發明之一蛋白質 或一胜肽(多胜肽),藉由偵測於一樣本(例如,血液、 組織)中之一抗體可診斷癌症。製備本發明蛋白質與胜肽 的方法如上所述》 如上所述,藉由測定介於抗MYBL2抗體的量與其在對 應控制組中的量之間的差異可執行癌症之診斷方法。若個 體之細胞或組織含有抗基因之表現產物(MYBL2)之抗體且 抗MYBL2抗體的量被測定大於在相較於其在正常控制組之 程度中的截斷值時,個體被懷疑遭受癌症。 在另一實施例中,本發明之診斷套組可包括本發明之 胜狀與結合至其之HLA分子1用抗原胜肽與腸分子偵 測抗原專-細胞毒殺性T淋巴球的方法已被建立(例如,Cglyceraldehyde-3-phosphate dehydrogenase)^ ^ ^ ^ White P Bu When compared with the normal control group, the degree of expression of the MYBL2 gene is increased or phase A / equivalent to the degree of cancer control group, the individual can be diagnosed as having cancer to be treated. The present invention also provides (1) a method of diagnosing or confirming whether a subject is suspected of having cancer to be treated, knife/and/or (11) selecting an individual to be treated for cancer, the method comprising the steps of · 201200525 a) measuring in cancer 钿In cells or tissues, the degree of expression of MYBL2 is obtained from individuals suspected of having cancer to be treated; b) the degree of expression of MYBL2 compared with the normal control group; the degree of performance of 〇若丑2 and the degree of normal control group The individual is diagnosed as having the cancer to be treated; and d) the individual who is selected or confirmed to be treated for cancer if the individual is expected to have the cancer to be treated in (4) c). Or 'this method may include the steps of: a) determining the extent of expression of MYBL2 in a cancer cell or tissue obtained from an individual suspected of having a cancer to be treated; b) comparing MYBL2 with a cancer control group Degree of performance; c) if the degree of expression of MYBL2 is similar or equal to the extent of the cancer control group, the individual is diagnosed as having cancer to be treated; and d) if the individual is diagnosed as having cancer to be treated in step c) Select or confirm the individual to be treated for cancer. The invention is also intended for use in a kit to determine that a subject is suffering from a cancer that can be treated with the MYBL2 multipeptide of the invention, which is also useful in assessing and/or monitoring the efficacy of cancer immunotherapy. Preferably, the cancer includes, but is not limited to, sputum tumor, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer, and esophageal cancer. More particularly, the kit preferably includes at least one reagent for detecting the degree of expression of the MYBL2 gene from an individual cancer cell, the reagents being selected from the group: (a) - the reagent for detecting the MYBL2 gene (b) - a reagent for detecting MYBL2 protein; and 57 201200525 (C) a reagent for detecting the biological activity of MYBL2 protein. Examples of a reagent suitable for detecting mRNA of the MYBL2 gene may include a nucleic acid which specifically binds or recognizes MYBL2 mRNA, for example, a nucleotide having a sequence partially complementary to one of the 2 mRNAs. These types of oligonucleotides are exemplified by primers and probes specific for 瞧2 mRNA. These kinds of raised nucleotides can be prepared according to methods well known in the art. If necessary, the reagent for detecting MYBL2 mRNA can be immobilized on a solid substrate (in addition, more than one reagent for detecting MYBL2 mRNA can be included in the kit. On the other hand, it is suitable for Examples of the reagent for detecting the MYBL2 protein include an antibody against the MYBL2 protein. The antibody may be a single plant or a plurality of strains. Further, any fragment or modification of the antibody (e.g., chihhc antibody, ScFv, Fab, F (ab) , ) 2, Fv, etc. can be used as a reagent as long as the fragment or modified antibody maintains the ability to bind to the MYBL2 protein. Methods for preparing these kinds of antibodies for protein detection are well known in the art, and Any method can be used in the present invention to prepare such an antibody and its equivalent e. Alternatively, the signal generating molecule can be labeled by direct ligation or an indirect labeling technique. Methods and detection of markers and marker antibodies The binding of antibodies to their targets is well known in the art, and any of the markers and methods can be used in the present invention. In addition, more than one is used for The reagent for measuring the MYBL2 protein can be included in the kit. The kit can contain more than one of the foregoing reagents. For example, a tissue sample obtained from an individual without cancer or suffering from cancer can be used as a useful control group test 5 58 201200525. The kit of the present invention may further comprise other materials required by the commercial or user and user perspective, including buffer solution, diluent clearing 15 / main shot needle, syringe and package insert with operational instructions for use (eg, f face, — etc.. These reagents or the like can be maintained in one of the following contents: Suitable containers include bottles, small glass bottles (~υ and test tubes. Containers can be formed from a variety of materials, such as glass Or plastic. In the present invention - when the reagent is a probe against the (four) heart test, the reagent may be immobilized on a solid substrate, such as a porous strip ("(8) to form at least - (four) positions. The measurement or zone of the porous strip can include a plurality of locations 'each containing a nucleic acid (probe). A test strip can also contain a negative and/or positive control set position. Alternatively, the position of the control set can be located The strip is separated by one strip. Full two &amp; - depending on the 'different detection bits may contain different amounts of immobilized nucleic acid' ie a higher amount in the first measurement position and a lower content in the subsequent Positional order. By adding the test sample, some positions of the detectable number are provided to provide a quantitative indication of the amount of MYBL2 mRNA present in the sample. The 1 position can be set to any suitable detectable shape and is generally across - the shape of the strip or dot of the width of the test strip. The kit of the present invention may further comprise a positive control group sample or a MYBL2 standard sample. By collecting the ship 2 positive samples and then analyzing their ugly 2 degree A positive control group sample of the invention is prepared. Alternatively, purified MYBL2 protein or polynucleic acid can be added to cells that do not exhibit 2 to form a positive sampie or MYBL2 standard sample. In the present invention, a purified MYBL2 may be a recombinant protein. The degree of _2 of the positive control group sample is, for example, greater than the cutoff value (cut〇ff vaiue). 59 201200525 In one embodiment, the invention further provides a diagnostic kit comprising a protein or a portion thereof protein having the ability to specifically recognize an antibody or fragment thereof of the invention. Examples of partial peptides of the protein of the present invention include polypeptides consisting of at least 8, preferably 15, more preferably 20 contiguous amino acids in the amino acid sequence of the protein of the present invention. Using one of the proteins of the present invention or a peptide (polypeptide), cancer can be diagnosed by detecting one of the antibodies (e.g., blood, tissue). The method of preparing the protein of the present invention and the peptide is as described above. As described above, the diagnosis method of cancer can be performed by measuring the difference between the amount of the anti-MYBL2 antibody and its amount in the corresponding control group. If the individual's cells or tissues contain an antibody against the gene expression product (MYBL2) and the amount of the anti-MYBL2 antibody is determined to be greater than the cut-off value in the extent of its normal control group, the individual is suspected of suffering from cancer. In another embodiment, the diagnostic kit of the present invention may comprise a method of the present invention and a method for detecting an antigen-cell cytotoxic T lymphocyte with an antigen peptide and an intestinal molecule for HLA molecule 1 bound thereto Build (for example,

Altman JD et al., Science. 1996, 274(5284): 94-6) 〇 因此,本發明之胜肽與HLA分子的複合物可應用至债測腫 瘤抗原專-細胞毒殺性T淋巴球的该測方法,藉此使早期 偵測癌症之復發及/或轉移為可能。此外,其可被用於適八 包含本發明胜肽為一活性成分之藥物的個體的薛選,或藥 物治療功效的評估。 、 特別是’根據已知方法(參見’例如M t謹JD吋Μ · ⑤ 60 201200525 jAltman JD et al., Science. 1996, 274(5284): 94-6) 〇 Therefore, the complex of the peptide of the present invention and the HLA molecule can be applied to the tumor cell antigen-cytotoxic T lymphocyte The method of measurement is used to enable early detection of recurrence and/or metastasis of cancer. Furthermore, it can be used for the selection of the individual, or the evaluation of the therapeutic efficacy of the drug, which comprises a drug comprising the peptide of the present invention as an active ingredient. , in particular ' according to known methods (see 'for example, M t JD吋Μ · 5 60 201200525 j

Science. 1996, 274(5284): 94_6),可製備放射標誌之 HLA分子與本發明胜肽之寡聚多聚(〇lig〇mermuitimer)複 合物,例如四聚體。伴隨使用複合物,可執行診斷,例如 藉由將來自被懷疑遭受癌症之個體的周邊血液淋巴球 (peripheral blood lymphocytes)中之抗原-胜肽專一細胞 毒殺性T淋巴球進行定量。 本發明更提供藉由使用此處敘述之胜肽抗原決定位之 用以評估個體之免疫反應的診斷方法或試劑。在本發明一 實施例中,如上述之HLA-A-2限制胜肽被使用為評估或預 測一個體之免疫反應的試劑。藉由將免疫抗原(i mmun〇gen) 與免疫活性細胞(immunocompetent)/;? 叩或 η. iro 接觸可誘導要被評估之免疫反應。在較佳實施例中,用以 評估一免疫反應的免疫活性細胞可選擇自周邊血液、周邊 血液淋巴球(PBL)、與周邊血液單核細胞(pBMc).中。收集或 分離此類免疫活性細胞的方法為本技術領域所熟知。在一 些實施例中,可導致抗原專一細胞毒殺性T淋巴球的產生 的任何試劑可使用為試劑’而細胞毒殺性T淋巴球辨認與 結合至胜肽抗原決定位。胜肽試劑必須不被使用為免疫抗 原。用於此類分析之分析系統包括相當新近之技術發展, 例如四聚體,對細胞内淋巴激素(lymph〇kines)之染色與干 擾素釋放分析或EL I SPOT分析。在較佳實施例中,要與胜 狀試劑接觸之免疫活性細胞可為包括樹突細胞之抗原呈現 細胞。 例如’本發明胜肽可使用於四聚體染色分析中以評估 61 201200525 為了抗原專一細胞毒殺性τ淋巴球存在之周邊血液單核細 胞,在暴露至腫瘤抗原或一免疫抗原後。HLA四聚體複合 物可被使用來直接顯現抗原專一細胞毒殺性τ淋巴球(參 見,例如 〇gg et al., Science 279: 2103-2106,1998; 與 Altman et al,Science 1 74 : 94-96,1 996 ),並測 定於周邊血液單核細胞之樣本中的抗原專一細胞毒殺性T 淋巴球族群的頻率。四聚體多聚體試劑,例如使用本發明 胜肽之四聚體試劑可如下被產生,如下所述。 在對應之HLA重鏈與冷2-微球蛋白存在下重新折疊結 合至HLA之胜肽,以產生三分子複合物。在複合物中,重 键之羧端為經生物素化於一預先設計進入蛋白質之位置。 之後將卵白素加至複合物以形成由三分子複合物與卵白素 (streptavidin)所組成之四聚體。藉由以螢光標誌卵白素 的方式’可使用四聚體來對抗原呈現細胞染色。之後可鑑 定細胞,例如藉由流式細胞技術。此類分析可被用於診斷 與預後(prognostic)目的。藉由此程序鑑定之細胞也可被 用於治療目的。Science. 1996, 274(5284): 94_6), can prepare a radiolabeled HLA molecule and an oligomeric poly(polyligomer) complex of the peptide of the present invention, such as a tetramer. Along with the use of the complex, a diagnosis can be performed, for example, by quantifying the antigen-peptide specific cell toxic T lymphocytes in peripheral blood lymphocytes from individuals suspected of suffering from cancer. The invention further provides diagnostic methods or reagents for assessing an individual's immune response by using the peptide epitopes described herein. In one embodiment of the invention, the HLA-A-2 restricted peptide as described above is used as an agent for assessing or predicting an immune response in a subject. The immune response to be assessed can be induced by contacting the immunizing antigen (i mmun〇gen) with immunocompetent cells/;? or η. iro. In a preferred embodiment, the immunocompetent cells used to assess an immune response are selected from peripheral blood, peripheral blood lymphocytes (PBL), and peripheral blood mononuclear cells (pBMc). Methods of collecting or isolating such immunocompetent cells are well known in the art. In some embodiments, any agent that results in the production of antigen-specific cytotoxic T lymphocytes can be used as a reagent&apos; and cytotoxic T lymphocytes recognize and bind to a peptide epitope. The peptide reagent must not be used as an immunoreactive antigen. Analytical systems for such assays include fairly recent technological developments such as tetramers, staining of lymphocytes in lymphocytes and interferon release assays or EL I SPOT assays. In a preferred embodiment, the immunocompetent cells to be contacted with the victer reagent may be antigen presenting cells including dendritic cells. For example, the peptide of the present invention can be used in a tetramer staining assay to evaluate 61 201200525 for peripheral blood mononuclear cells in the presence of antigen-specific cytotoxic lytic lymphocytes after exposure to a tumor antigen or an immune antigen. HLA tetrameric complexes can be used to directly visualize antigen-specific cytotoxic lytic lymphocytes (see, for example, 〇gg et al., Science 279: 2103-2106, 1998; and Altman et al, Science 1 74: 94- 96,1 996), and measures the frequency of antigen-specific cytotoxic T lymphocyte populations in samples of peripheral blood mononuclear cells. A tetrameric multimeric reagent, for example, a tetrameric reagent using the peptide of the present invention can be produced as follows, as described below. The peptide that binds to HLA is refolded in the presence of the corresponding HLA heavy chain and cold 2-microglobulin to produce a three molecule complex. In the complex, the carboxy terminus of the heavy bond is biotinylated at a pre-designed location into the protein. The avidin is then added to the complex to form a tetramer composed of a tri-molecular complex and streptavidin. The antigen can be stained with cells by using tetramers in the form of fluorescently labeled avidin. The cells can then be identified, for example by flow cytometry. Such analysis can be used for diagnostic and prognostic purposes. Cells identified by this procedure can also be used for therapeutic purposes.

本發明也提供評估免疫收回反應(immune recaH responses)之試劑(參見’例如 Bertoni etaL,J. Clin.The invention also provides reagents for assessing immune recaH responses (see ' for example, Bertoni etaL, J. Clin.

Invest. 100: 503-513, 1997 and Penna et aL, J Exp. Med. 174: 1565-1570,1991) ’其包括本發明之胜肽。例如, 為了抗原-專一細胞毒殺性T淋巴球的存在,使用特定專一 胜肽’可分析來自具有要被治療之癌症之個體的病患pBMC 樣本。藉由培養PBMC與以本發明胜肽刺激細胞可評估含單 62 ⑤ 201200525 核細胞之血液樣本。在適合之培養期間後,例如為了細胞 毒殺性τ淋巴球活性,分析經擴張之細胞族群。 胜肽也可使用為評估—疫苗功效之試劑。使用例如任 -上述方法可分析獲自1 _免疫抗原接種之病患的 PBMC。病患為、經HLA分型,且選擇辨認表現於病患中之對 偶基因(allelespecific)分子的胜肽抗原試劑以分析。藉 由於PBMC樣本中之抗原決定位_專一細胞毒殺性τ淋巴球 的存在,可才日出疫之免疫抗原性(丨關⑽叩印丨c丨^ )。Invest. 100: 503-513, 1997 and Penna et al, J Exp. Med. 174: 1565-1570, 1991) 'which includes the peptide of the present invention. For example, for the presence of antigen-specific cytotoxic T lymphocytes, a patient-specific pBMC sample from an individual having a cancer to be treated can be analyzed using a specific specific peptide. Blood samples containing singular 62 5 201200525 nucleated cells can be evaluated by culturing PBMC and stimulating cells with the peptide of the present invention. The expanded cell population is analyzed, for example, for cytotoxic tau lymphocyte activity during a suitable culture period. The peptide can also be used as an agent for evaluating the efficacy of the vaccine. The PBMC obtained from the patient vaccinated with the 1-immuno antigen can be analyzed using, for example, any of the above methods. The patient is, HLA-typed, and selected to recognize the peptide antigen reagents present in the allele specific molecule of the patient for analysis. By the presence of the antigenic determinant in the PBMC sample _ specific cell toxic τ lymphocytes, it is only possible to immunize antigenicity (丨(10)叩印丨c丨^).

本發明之胜肽也可用來製造抗體,使用本技術領域已 熟知的技術(參見,例如,CURRENT PR0T0C0LS IN IMMUNOLOGY, Wi1ey/Greene, NY ; and Antibodies A Laboratory Manual, Harlow and Lane, Cold Spring Harbor Laboratory Press,1 989 ),其可有效作為診斷或 監測癌症之試劑。此類抗體可包括辨認於HLA分子背景中 之胜肽的那些,即,結合至胜肽-MHC複合物的抗體。 或者,本發明也提供一些用途,其之一些為此處所述。 例如’本發明提供診斷或偵測以MYBL2免疫活性多胜肽之 表現為特徵的一疾病。這些方法包含測定於一生物樣本中 之MYBL2 HLA結合胜肽的表現或MYBL2 HLA結合胜肽與HLA c 1 ass I分子之一複合物。藉由以對於胜肽或複合物之結 合伙伴(binding partner)分析可測定或偵測一胜肽之表 現或胜肽與HLA c 1 ass I分子之複合物。在一較佳實施例 中,對於胜肽或複合物之結合伙伴為一抗體其辨認且專一 結合至胜肽。藉由使用MYBL2引子之標準PCR放大步驟也 63 201200525 可測試於一生物樣本’例如一腫瘤切片中之MYBL2的表 現。腫瘤表現之例子於此被呈現且示範之用於MYBL2放大 的條件與引子的更進一步揭露可被發現於W02003/27322 中〇 較佳為’診斷方法包含將分離自一個體的生物樣本與 專一於MYBL2 HLA結合胜肽之一試劑接觸以偵測於生物樣 本中之MYBL2 HLA結合胜肽的存在。如此處所使用“接觸” 意扣以有效接近試劑方式放置生物樣本且在適合之例如, 濃度、溫度、時間 '離子強度條件下,以允許介於試劑與 存在生物樣本中之MYBL2 HLA結合胜肽的專一互相作用。 一般而§,將試劑接觸生物樣本之條件為熟悉此技藝人士 所知之條件以促進介於分子及於生物樣本中之其同類物 (cognate)(例如,一蛋白質與其受體同類物、一抗體與其 蛋白質抗原同類物、一核酸與其互補序列同類物)之間的 專一互相作用。促進介於分子與其同類物之間的專一互相 作用的不範條件敘述於L〇wetal所提出之U. s. PatentThe peptide of the present invention can also be used to make antibodies using techniques well known in the art (see, for example, CURRENT PR0T0C0LS IN IMMUNOLOGY, Wi1ey/Greene, NY; and Antibodies A Laboratory Manual, Harlow and Lane, Cold Spring Harbor Laboratory Press , 1 989 ), which is effective as a reagent for diagnosing or monitoring cancer. Such antibodies may include those recognized as peptides in the context of HLA molecules, i.e., antibodies that bind to the peptide-MHC complex. Alternatively, the invention also provides some uses, some of which are described herein. For example, the present invention provides a diagnosis or detection of a disease characterized by the expression of a MYBL2 immunoreactive multi-peptide. These methods comprise assaying the expression of a MYBL2 HLA-binding peptide in a biological sample or a complex of a MYBL2 HLA-binding peptide with an HLA c 1 ass I molecule. The expression of a peptide or the complex of a peptide with an HLA c 1 ass I molecule can be determined or detected by analysis of a binding partner for a peptide or complex. In a preferred embodiment, the binding partner for the peptide or complex is an antibody that recognizes and specifically binds to the peptide. The standard PCR amplification step using the MYBL2 primer is also 63 201200525. The performance of MYBL2 in a biological sample, such as a tumor slice, can be tested. Examples of tumor manifestations The further disclosure of conditions and primers for MYBL2 amplification presented and demonstrated herein can be found in WO 2003/27322. Preferably, the diagnostic method comprises biological samples separated from one body and specific ones. One of the MYBL2 HLA binding peptides is contacted to detect the presence of the MYBL2 HLA binding peptide in the biological sample. As used herein, "contacting" means placing a biological sample in an effective proximity reagent and, for example, at a concentration, temperature, time 'ion strength, for example, to allow binding of the peptide to the MYBL2 HLA in the biological sample. Specific interaction. In general, §, the conditions under which the agent is contacted with the biological sample are those known to those skilled in the art to promote cognate between the molecule and the biological sample (eg, a protein and its receptor analog, an antibody) A specific interaction with its protein antigen congener, a nucleic acid and its complementary sequence congener. U. s. Patent proposed by L〇wetal to promote the specific interaction between the molecule and its congeners.

No. 5, 1 08, 921。 可在2/7 H 或“ 汁0之一或兩者執行本發明之診 斷方法。因此,在本發明中生物樣本可位於加或 wiro中例如,生物樣本可為一 y λ h 組織且專一於 MYBL2免疫活性多胜肽的試劑可被用來偵測於組織中此類 分子的存在。或者,可h⑽收集或分離生物樣本(例 如血液樣本、腫瘤切片、組織萃取物)。在一特別較佳實 施例中,±物樣本可為一含細胞縣,更佳為一樣本含有 64 201200525 收集自要被診斷或測試之個體的腫瘤細胞。 或者,藉由一方法可執行診斷,此方法藉由以螢光素 (fluorescein)標諸HLA多聚複合物染色允許抗原-專一 T 細胞的直接定量(例如,A1 tman,J. D. et al., 1996, Science 274: 94; Altman, J. D. e t a 1. , 1 993,Proc. Natl. Acad. Sci. USA 90: 1 0330 )。也已提供細胞内淋巴激素 (lymphokines)之染色與干擾素τ釋放分析或ELISP0T分 析。四聚體多聚體染色、細胞内淋巴激素染色與ELI SPOT 分析皆顯露比一般分析靈敏至少多10倍(Mural i-Krishna, K. et al., 1998, Immunity 8: 177; Lalvani, A. et al., 1 997, J. Exp. Med. 1 86: 859; Dunbar, P. R. et al., 1 998, Curr. Biol. 8 : 413 ;)。也可使用五聚體(例如,US 2004-209295A )、右聚體(dextramer)(例如,w〇 02/072631 )、鏈聚體(streptamer)(例如,Nature medicine 6. 631-637 (2002))。 例如’在一些實施例中,本發明提供診斷或評估被投 予本發明至少一 MYBL2胜肽之個體的免疫反應的方法,方 法包括步驟: (a )在適合誘導專一於免疫原之細胞毒殺性了淋巴球 的條件下’將免疫原與免疫活性細胞接觸; (b) 偵測或測定於步驟(a)中所誘導之細胞毒殺性j 淋巴球的誘導程度;以及 (c) 使個體之免疫反應與細胞毒殺性τ淋巴球的誘導 私度互相關連。 65 201200525 在本發明中,免疫原為(a)擇自序列辨識號:2至4之胺基 酸序列中的MYBL2胜肽、具有此種胺基酸序列的胜肽與具 有此種胺基酸序列於其中已被以丨、2或更多胺基酸取代所 修飾的胜肽的至少一個。於期間,適合誘導免疫原專一之 細胞毒殺性T淋巴球的條件為本技術領域所熟知。例如, 可in vitro培養免疫活性細胞在免疫原存在下以誘導免疫 原專一之細胞毒殺性T淋巴球。為了誘導免疫原專一之細 胞毒殺性T淋巴球,可加入任何刺激因子於細胞培養物 中。例如,IL-2為細胞毒殺性τ淋巴球誘導之較佳刺激因 〇 在一些實施例中,可在治療前、期間及/或後執行監測 或評估要被以胜肽癌症治療之個體的免疫反應的步驟。一 般而s ’在癌症治療的程序中,一再地將免疫原性胜肽投 予至要被治療的個體。例如’可每週投予免疫原性胜肽達 3-1 0週。因此,在癌症治療程序期間,可評估或監測個體 之免疫反應。或者,對於癌症治療之免疫反應的評估或監 測的步驟可在治療程序完成時。 根據本發明’當與控制組相較時,經增強之免疫原專 一之細胞毒殺性T淋巴球的誘導指出要被評估或診斷之個 體免疫性地對已被投予之免疫原反應。用以評估免疫反應 之適合的控制組可包括’例如當免疫活性細胞未與胜肽接 觸或與具有除了任何MYBL2胜肽之胺基酸序列的控制組胜 狀(例如’隨機胺基酸序列)接觸時的細胞毒殺性T淋巴 球的誘導程度。 66 201200525 在一較佳實施例中,藉由將介於投予至個體之各個免 疫原間的免疫反應進行比較,以序列專一方式來評估個體 之免疫反應。特別是’即使當將MYBL2胜肽之一些種類的 混合物投予至個體時,依據胜肽,免疫反應可成多樣化。 在此例子中,藉由將介於各個胜肽間的免疫反應進行比 較,可確認出對於其個體顯示較高反應之胜肽。 XI.抗體 本發明提供抗體其結合至本發明胜肽,較佳抗體專一 結合至本發明胜肽且不會結合(或微弱結合)非本發明胜 狀。或者抗體結合本發明胜肽與其同源物(h〇m〇1〇gs)。 抗本發明胜肽之抗體可提供用途於癌症診斷與預後分 析及成像方法(i mag i ng me thodo 1 ogi es )。相似地,對於在 癌症病患中也表現或過度表現MYBL2程度而言,此類抗體 可提供使用於其他癌症之治療、診斷,及/或預後。此外, 細胞内表現之抗體(例如,單鏈抗體)在治療與之 表現相關的癌症中為治療性有效的,與MYBL2之表現相關 的癌症例子包括,但不限於,乳癌、子宮頸癌、大腸直腸 癌、食道癌、月癌、溺沒型胃癌、淋巴癌、神經母細胞瘤、 胰臟癌。 本發明也提供MYBL2蛋白質(序列辨識號:6 )或其片 段之偵測及/或定量的各種免疫活性分析,MYBL2蛋白質(序 列辨識號.6)或其片段包括具有擇自序列辨識號:2至4 中之胺基酸序列之多胜肽。此類分析可包括一或多個具辨 認及結合MYBL2蛋白質或其片段之能力之抗财肌2抗體為 67 201200525 適當的。在本發明内容中,與MYBL2多胜肽結合之抗MYBL2 抗體’較佳為辨認一多胜肽,此多胜肽為具有擇自序列辨 識號.2至4中的胺基酸序列。以抑制測試(丨n h i b i t i ο η 1: e s t) 可確認抗體之結合專一性。其為,在具有擇自序列辨識號: 2至4中之胺基酸序列之任何片段多胜肽存在下,當介於 要被分析之抗體與全長之MYBL2多胜肽之間的結合被抑制 時’其顯示此抗體專一結合至片段。在本發明内容中,在 包括’但不限於放射免疫分析(radi〇immunoassayS)、免疫 色層分析技術(immuno-chromatgraph technique)、酵素連 結免疫吸附分析(enzyme-linked immunosorbent assays, ELISA)、酵素連結免疫勞光分析(enZyme_ linked immunofluorescent assays,ELIFA)等之多種形式本技術 領域熟知之免疫分析形式中執行此類免疫分析。 本發明之關於免疫’但非抗體分析也可包括T細胞致 免疫性分析(immun〇genic ity assay)(抑制或刺激)與主 要組織相容性複合物(major hi stocompatibi 1 ity complex) 結合分析。此外,本發明也提供具偵測表現MYBL2之癌症 之能力的免疫成像分析,包括,但不限於使用本發明之經 標諸的抗體的放射顯像成像(radio scintigraphic waging)方法。此類分析在mYBL2表現之癌症的偵測、監 控與預後的為臨床有效的,MYBL2表現之癌症,其例子包 括’但不限於’乳癌、子宮頸癌、大腸直腸癌、食道癌、 月癌、瀰漫型胃癌、淋巴癌、神經母細胞瘤、胰臟癌。 本發明提供結合至本發明胜肽的抗體。本發明抗體可 68 201200525 使用於任何形式中, 例如單株或多株抗體,且包括獲得自No. 5, 1 08, 921. The diagnostic method of the present invention can be performed at one or both of 2/7 H or "Juice 0. Thus, in the present invention, the biological sample can be located in plus or wiro. For example, the biological sample can be a y λ h tissue and is specific to The MYBL2 immunoreactive peptide peptide can be used to detect the presence of such molecules in the tissue. Alternatively, the biological sample (e.g., blood sample, tumor section, tissue extract) can be collected or isolated by h(10). In an embodiment, the ± sample may be a cell-containing county, more preferably the same as the tumor cell collected from the individual to be diagnosed or tested 64 201200525. Alternatively, the diagnosis may be performed by a method by Fluorescence of fluorescein labeled HLA polyplex allows direct quantification of antigen-specific T cells (eg, A1 tman, JD et al., 1996, Science 274: 94; Altman, JD eta 1., 1 993 , Proc. Natl. Acad. Sci. USA 90: 1 0330 ). Intracellular lymphokines staining and interferon tau release assay or ELISPOT analysis have also been provided. Tetramer multimer staining, intracellular lymphokines Dyeing and ELI SPOT analysis revealed at least 10 times more sensitivity than general analysis (Mural i-Krishna, K. et al., 1998, Immunity 8: 177; Lalvani, A. et al., 1 997, J. Exp. Med. 1 86: 859; Dunbar, PR et al., 1 998, Curr. Biol. 8 : 413 ;) Pentamers (for example, US 2004-209295A), dextramers (for example, w〇) can also be used. 02/072631), a streptamer (eg, Nature medicine 6. 631-637 (2002)). For example, 'in some embodiments, the invention provides for the diagnosis or evaluation of at least one MYBL2 peptide administered to the invention. A method for the individual's immune response, the method comprising the steps of: (a) contacting the immunogen with an immunologically active cell under conditions suitable for inducing a lymphocyte specific to the immunogen; (b) detecting or measuring The degree of induction of cytotoxic j lymphocytes induced in step (a); and (c) correlating the individual's immune response with the induction of cellular toxic lymphocytes. 65 201200525 In the present invention, the immunogen For (a) the MYBL2 peptide in the amino acid sequence of sequence identification number: 2 to 4, with such an amine The peptide of the acid sequence is at least one of a peptide having such an amino acid sequence modified therein by substitution with hydrazine, 2 or more amino acids. Conditions suitable for inducing immunogenic cytotoxic T lymphocytes during the period are well known in the art. For example, immunocompetent cells can be cultured in vitro in the presence of an immunogen to induce immunogenic cytotoxic T lymphocytes. To induce immunogen-specific cytotoxic T lymphocytes, any stimulating factor can be added to the cell culture. For example, IL-2 is a preferred stimulator of cytotoxic tau lymphocyte induction. In some embodiments, monitoring or assessing immunity of an individual to be treated with a peptide cancer can be performed before, during, and/or after treatment. The step of the reaction. In general, in the course of cancer treatment, the immunogenic peptide is repeatedly administered to the individual to be treated. For example, the immunogenic peptide can be administered weekly for 3-1 0 weeks. Thus, an individual's immune response can be assessed or monitored during the cancer treatment procedure. Alternatively, the step of assessing or monitoring the immune response to cancer treatment may be at the completion of the treatment procedure. According to the present invention, the induction of the enhanced immunogen-specific cytotoxic T lymphocytes indicates that the individual to be evaluated or diagnosed immunologically responds to the administered immunogen when compared with the control group. A suitable control set for assessing an immune response can include, for example, 'when the immunocompetent cells are not in contact with the peptide or with a control group having an amino acid sequence other than any MYBL2 peptide (eg, a 'random amino acid sequence') The degree of induction of cytotoxic T lymphocytes upon exposure. 66 201200525 In a preferred embodiment, an individual's immune response is assessed in a sequence-specific manner by comparing the immune responses administered between the individual immunogens administered to the individual. In particular, even when a mixture of some species of MYBL2 peptide is administered to an individual, the immune response can be diversified depending on the peptide. In this example, by comparing the immune responses between the individual peptides, it was confirmed that the peptide which showed a higher response to the individual was confirmed. XI. Antibodies The present invention provides antibodies which bind to the peptides of the present invention. Preferably, the antibodies bind specifically to the peptide of the present invention and do not bind (or weakly bind) to the present invention. Alternatively, the antibody binds to the peptide of the invention and its homolog (h〇m〇1〇gs). The antibody against the peptide of the present invention can be used for cancer diagnosis and prognosis analysis and imaging methods (i mag i ng me thodo 1 ogi es ). Similarly, such antibodies may provide for the treatment, diagnosis, and/or prognosis of other cancers for the extent that they also exhibit or overexpress MYBL2 in cancer patients. In addition, antibodies that are expressed intracellularly (eg, single-chain antibodies) are therapeutically effective in treating cancers associated with their performance, and examples of cancers associated with the expression of MYBL2 include, but are not limited to, breast cancer, cervical cancer, large intestine Rectal cancer, esophageal cancer, lunar cancer, sputum-type gastric cancer, lymphoma, neuroblastoma, pancreatic cancer. The present invention also provides various immunological activity assays for detecting and/or quantifying MYBL2 protein (SEQ ID NO: 6) or a fragment thereof, MYBL2 protein (SEQ ID NO: 6) or a fragment thereof comprising the selected sequence identification number: 2 Multi-peptide of the amino acid sequence up to 4. Such assays may include one or more anti-fighting muscle 2 antibodies that recognize and bind to the MYBL2 protein or a fragment thereof as 67 201200525. In the context of the present invention, an anti-MYBL2 antibody&apos; that binds to a MYBL2 polypeptide is preferably a multi-peptide which is an amino acid sequence having the sequence identification number. The binding specificity of the antibody can be confirmed by the inhibition test (丨n h i b i t i ο η 1: e s t). In the presence of any fragment of the peptide having the amino acid sequence selected from the sequence identification numbers: 2 to 4, the binding between the antibody to be analyzed and the full-length MYBL2 polypeptide is inhibited. When it shows that this antibody binds specifically to the fragment. In the context of the present invention, including but not limited to radioimmunoassayS, immuno-chromatgraph techniques, enzyme-linked immunosorbent assays (ELISA), enzyme linkages Various forms of enZyme-linked immunofluorescent assays (ELIFA) and the like perform such immunoassays in immunoassay formats well known in the art. The immunological but non-antibody analysis of the present invention may also include a T cell immunogenic assay (inhibition or stimulation) in combination with a major histocompatibility complex. Furthermore, the present invention also provides immunoimaging assays having the ability to detect cancers that exhibit MYBL2, including, but not limited to, radio scintigraphic waging using the labeled antibodies of the present invention. Such analysis is clinically effective in detecting, monitoring, and prognosing cancers in mYBL2, and MYBL2 is a cancer, examples of which include, but are not limited to, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, and monthly cancer. Diffuse gastric cancer, lymphoma, neuroblastoma, pancreatic cancer. The invention provides antibodies that bind to the peptides of the invention. The antibody of the present invention can be used in any form, for example, single or multiple antibodies, and includes obtained from

人源化抗體。 使用為一杬原以獲得一抗體之本發明胜肽可來自任何 乳動物’例如,人類、小鼠或 人類來源胜肽可獲得自此處揭 動物種類,但較佳為來自哺乳動物 大鼠,更佳為來自一人類。人類來 露之核苷酸或胺基酸序列。 根據本發明’使用為免疫抗原之胜肽可為一完整之蛋 白質或蛋白質之部分胜肽。部分胜肽可包括,例如,本發 明之一胜肽之胺基(N)端或羧基(c)端片段。 此處’一抗體被定義為一蛋白質其與MYBL2胜肽之全 長或片段反應。在一較佳實施例中,本發明之抗體辨認 MYBL2片段胜肽’其具有擇自序列辨識號:2至4中的胺基 酸序列。合成寡胜肽的方法為本技術領域所熟知。在合成 後,在使用為免疫抗原(immun〇gen)前,胜肽可視需要被純 化。在本發明中’寡胜肽(例如9或10員)可與載體結合 或連結以增強致免疫性。鑰孔血藍蛋白(Keyhole-1 impet hemocyanin,KLH)被熟知為載體。結合鑰孔血藍蛋白與胜 狀的方法為本技術領域所熟知。 或者’可將編碼出本發明一胜肽或其片段的一基因插 入一已知的表現載體,其之後被轉形至一宿主細胞,如於 此所敘述。藉由任何標準方法,所需之胜肽或其片段可自 估主細胞之外部或内部被重新獲得,且之後可被使用為一 69 201200525 抗原。或者,可將表現胜狀之整個細胞或其細胞 一化學合成胜肽使用為抗原。 5 可以抗原將任何喝乳動物進行免疫,但較佳為考慮盘 用來細胞融合之親代細胞的相容性。一般而言,使用懷齒 目(Rodentia)、兔形目(Ug〇m〇rpha)或靈長目 (Pnmates)。噶齒目的動物包括,例如小鼠、大鼠與倉鼠。 兔形目的動物包括,例如兔子。$長目的動物包括,二 狹鼻類(舊世界猴)猴+ ’例如馬來狼 fas⑽laris)、㈣(rhesus _key)、㈣拂(咖㈣ bab〇on)與黑猩獲(chimpanzees)。 以抗原免疫動物之方法為本技術領域所熟知。抗原之 腹腔内注射(intraperit〇neal injecti〇n)或皮下注射 (subcutaneous injection)為免疫哺乳動物之一標準方 法。更特別地,可將抗原稀釋或懸浮於一適合量的磷酸鹽 緩衝/合液 '生理食鹽水等。若需要,可將抗原懸浮液與適 合量之標準佐劑,例如佛氏完全佐劑(Freund,s c〇mplete adjuvant)混合,製成乳狀液(emulsi〇n)並且之後投予至哺 乳動物。較佳為其之後投予與適合量之佛氏不完全佐劑 (Freund s incomplete adjuvant)混合的抗原,每 4 至 21 天。也可使用一適合之載體來免疫。於上述免疫後,藉由 為了增加所需之抗體量,以標準方法檢驗血清。 藉由自被檢驗以增加於血清中之所需抗體的經免疫動 物收集血_液且藉由以任何一般方法自血液分離血清,可製 備抗本發明胜狀之多株抗體。多株抗體可包括含多株抗體 70 201200525 的血清,與含自血清分離之多株抗體的部分(fracti〇n)。 例如使用與本發明胜肽結合之親合管柱且更進一步使用蛋 白質A或蛋白質G管柱純化此部分,可自僅辨認本發明胜 肽之部分純化免疫球蛋白G或μ。 為了製備單株抗體,如上所述自經以抗原免疫並確認 於血清中所需抗體之增加程度的哺乳動物收集免疫細胞且 使免疫細胞遭遇細胞融合。用來細胞融合之免疫細胞,較 佳為獲自脾臟。其他要被與上述免疫細胞融合之親代細胞 包括,例如,哺乳動物之骨髓瘤(mye丨〇ma),且較佳為具有 以藥物篩選之融合細胞之獲得特性的骨髓瘤細胞。 根據已知方法,例如Milstein et al. (Galfre and Milstein,Methods EnZymol 73: 3-46 ( 1981 ))的方法, 可將上述免疫細胞與骨髓瘤細胞融合。 獲自細胞融合之產生的融合瘤,藉由將它們培養於標 準篩選培養基,例如HAT培養基(含亞黃嘌呤 (hypoxanthine)、氨蝶呤(amin〇pterin)和胸腺嘧啶 (thynudine)之培養基),可被篩選。通常持續細胞培養於 HAT培養基數天至數週,_間為允許除了所需融合瘤外之 其他細胞(非融合細胞)死亡。之後,執行標準限制稀釋 以筛選並複製產生所需抗體之融合瘤。 除了於其中為了製備融合瘤、以一抗原免疫一非人類 動物的上述方法,可以胜狀、表現胜肽之細胞或其細胞萃 取物“ 免疫人類淋巴細胞,例如被別病毒感染的 那些。之後,將經免疫之淋巴細胞與具不明確分裂能力之 201200525 來自人類之骨髓瘤,例如U266融合,以產生一產生所需人 類抗體之融合瘤’所需之人類抗體可與可被獲得之胜肽結 合(Unexamined Published Japanese Patent ApplicationHumanized antibody. The peptide of the present invention which is used as a monoclonal antibody to obtain an antibody may be derived from any dairy animal 'for example, a human, mouse or human derived peptide may be obtained from a mammalian species, but preferably from a mammalian rat. Better from a human being. Human nucleotides or amino acid sequences. The peptide used as an immunizing antigen according to the present invention may be a complete protein or a partial peptide of a protein. The partial peptide may include, for example, an amine (N) terminal or a carboxyl (c) terminal fragment of one of the peptides of the present invention. Here, an antibody is defined as a protein which reacts with the full length or fragment of the MYBL2 peptide. In a preferred embodiment, the antibody of the invention recognizes the MYBL2 fragment peptide which has the amino acid sequence selected from Sequence Numbers: 2 to 4. Methods of synthesizing oligopeptides are well known in the art. After synthesis, the peptide can be purified as needed prior to use as an immunizing antigen. In the present invention, an oligopeptide (e.g., 9 or 10 members) can be bound or linked to a carrier to enhance immunogenicity. Keyhole-1 impet hemocyanin (KLH) is known as a vector. Methods for binding keyhole limpet hemocyanin and triumph are well known in the art. Alternatively, a gene encoding a peptide of the present invention or a fragment thereof can be inserted into a known expression vector, which is then transformed into a host cell, as described herein. The desired peptide or fragment thereof can be re-acquired externally or internally to the host cell by any standard method and can then be used as a 69 201200525 antigen. Alternatively, the entire cell expressing a victory or a cell thereof, a chemically synthesized peptide, can be used as an antigen. 5 Any animal that is drinking milk can be immunized with an antigen, but it is preferred to consider the compatibility of the parental cells used for cell fusion. In general, use Rodentia, Ug〇m〇rpha or Pnmates. Animals of the caries include, for example, mice, rats, and hamsters. Rabbit-shaped animals include, for example, rabbits. $long-eyed animals include, two narrow-nose (old world monkey) monkeys + 'such as Malay wolf fas (10) laris), (four) (rhesus _key), (four) 拂 (ca (4) bab〇on) and chimpanzees. Methods for immunizing animals with antigens are well known in the art. Intraperital injection (intraperit〇neal injecti〇n) or subcutaneous injection is a standard method for immunizing mammals. More specifically, the antigen may be diluted or suspended in a suitable amount of phosphate buffer/mixed liquid 'physiological saline or the like. If desired, the antigen suspension may be mixed with a suitable amount of a standard adjuvant, such as Freund's complete adjuvant (Freund, s c〇mplete adjuvant) to prepare an emulsion (emulsi〇n) and then administered to the mammal. Preferably, it is administered followed by an antigen mixed with a suitable amount of Freunds incomplete adjuvant every 4 to 21 days. A suitable vector can also be used for immunization. After the above immunization, serum was tested by standard methods in order to increase the amount of antibody required. The multi-strain antibody against the present invention can be prepared by collecting blood from an immunologically active animal that has been tested to increase the desired antibody in serum and by separating the serum from the blood by any general method. A plurality of antibodies may include a serum containing a plurality of antibodies 70 201200525, and a portion containing a plurality of antibodies isolated from serum (fracti〇n). For example, using an affinity column coupled to a peptide of the present invention and further purifying the portion using a protein A or protein G column, the immunoglobulin G or μ can be purified from only a portion of the peptide of the present invention. To prepare a monoclonal antibody, the mammalian cells are collected from the mammals which have been immunized with the antigen and confirmed to the extent of the desired antibody in the serum as described above and subject the immune cells to cell fusion. The immune cells used for cell fusion are preferably obtained from the spleen. Other parental cells to be fused with the above-described immune cells include, for example, myomas of mammals, and preferably myeloma cells having acquired characteristics of drug-selected fused cells. The above immune cells can be fused with myeloma cells according to known methods, for example, by Milstein et al. (Galfre and Milstein, Methods EnZymol 73: 3-46 (1981)). Fusion tumors obtained from cell fusion by culturing them in standard screening media, such as HAT medium (medium containing hypoxanthine, amin〇pterin, and thynudine), Can be screened. Usually, the cells are continuously cultured in HAT medium for several days to several weeks, and _ between them is to allow death of other cells (non-fused cells) other than the desired fusion tumor. Thereafter, standard limiting dilutions are performed to screen and replicate the fusion tumors that produce the desired antibodies. In addition to the above method in which a non-human animal is immunized with an antigen in order to prepare a fusion tumor, the cell which is capable of winning, expressing the peptide or its cell extract "immunizes human lymphocytes, such as those infected with another virus. Thereafter, The immunized lymphocytes are fused with human myeloma, such as U266, with a non-clear cleavage ability to produce a fusion tumor that produces the desired human antibody. The desired human antibody can be combined with the peptide that can be obtained. (Unexamined Published Japanese Patent Application

No. Sho 63-17688)。 將所獲得之融合瘤之後轉植進入小鼠之腹腔且萃取腹 水。可純化所獲得之單株抗體,藉由例如硫酸錄沉澱、一 蛋白質A或蛋白質G管柱、DEAE離子交換色層分析或一與 本發明胜肽結合之親和管柱。本發明抗體不只可被使用來 純化與偵測本發明胜肽,也可作為本發明胜肽之促進劑與 拮抗劑的候選物。 或者,一產生抗體之免疫細胞,例如一經免疫之淋巴 細胞可藉由一致癌基因以永生且被使用來製備單株抗體。 也可使用基因工程技術來重組製備因此獲得之單株抗 體(參見,例如 Borrebaeck and Larrick,TherapeuticNo. Sho 63-17688). The obtained fusion tumor was then transferred into the abdominal cavity of the mouse and the ascites was extracted. The obtained monoclonal antibody can be purified by, for example, precipitation by sulfuric acid, a protein A or protein G column, DEAE ion exchange chromatography or an affinity column coupled to the peptide of the present invention. The antibody of the present invention can be used not only to purify and detect the peptide of the present invention, but also as a candidate for the promoter and antagonist of the peptide of the present invention. Alternatively, an antibody-producing immune cell, e.g., an immunized lymphocyte, can be immortalized by a consensus oncogene and used to prepare a monoclonal antibody. Genetic engineering techniques can also be used to recombinantly prepare the individual antibodies thus obtained (see, for example, Borrebaeck and Larrick, Therapeutic

Monoclonal Antibodies, published in the UnitedMonoclonal Antibodies, published in the United

Kingdom by MacMillan Publishers LTD ( 1990))。例如, 一編碼出抗體的DNA可自一免疫細胞,例如產生抗體之— 融合瘤或一經免疫的淋巴細胞被複製,插入一適合之載 體,且引入一宿主細胞以製備重組抗體。本發明也提供如 上述製備之重組抗體。Kingdom by MacMillan Publishers LTD (1990)). For example, a DNA encoding an antibody can be replicated from an immune cell, e.g., an antibody-producing tumor or an immunized lymphocyte, inserted into a suitable vector, and introduced into a host cell to produce a recombinant antibody. The present invention also provides a recombinant antibody prepared as described above.

此外,本發明抗體可為一抗體之片段或經修飾之抗 體’只要其結合一或多個本發明之胜肽。例如,抗體片段 可為Fab、F(ab,)2、Fv或單鏈Fv (scFv),於其中來自重 與輕鏈的Fv片段藉由合適的連結器來連接(Hust〇n e1: aI 72 ⑤ 201200525Furthermore, the antibody of the present invention may be a fragment of an antibody or a modified antibody as long as it binds one or more of the peptides of the present invention. For example, the antibody fragment can be a Fab, F(ab,)2, Fv or single-chain Fv (scFv) in which the Fv fragments from the heavy and light chains are joined by a suitable linker (Hust〇n e1: aI 72 5 201200525

Proc Natl Acad Sci USA 85: 5879-83 ( 1988))。更特別 是’藉由以酵素例如木瓜酵素或胃蛋白酶處理抗體可產生 抗體片段。或者,編碼出抗體之基因可被構築、插入一表 現載體且表現於一適合的宿主細胞中(參見,例如,C〇 et al. , J Immunol 152: 2968-76 ( 1 994); Better and Horwitz, Methods Enzymol 178: 476-96 (1989); Pluckthun and Skerra, Methods Enzymol 178: 497-515 (1989); Lamoyi, Methods Enzymol 121: 652-63 ( 1 986); Rousseaux et al., Methods Enzymol 121: 663-9 (1986); Bird and Walker,Proc Natl Acad Sci USA 85: 5879-83 (1988)). More specifically, antibody fragments can be produced by treating antibodies with enzymes such as papain or pepsin. Alternatively, the gene encoding the antibody can be constructed, inserted into a performance vector and displayed in a suitable host cell (see, for example, C〇 et al., J Immunol 152: 2968-76 (1 994); Better and Horwitz , Methods Enzymol 178: 476-96 (1989); Pluckthun and Skerra, Methods Enzymol 178: 497-515 (1989); Lamoyi, Methods Enzymol 121: 652-63 (1 986); Rousseaux et al., Methods Enzymol 121: 663-9 (1986); Bird and Walker,

Trends Biotechnol 9: 132-7 (1991))。 藉由與各種分子’例如聚乙二醇(p〇lyethylene glycol, PEG)結合可修飾抗體。本發明提供此類經修飾之 抗體。藉由化學修飾一抗體可獲得經修飾之抗體。這些修 飾方法為本技術領域中所常見。 或者,本發明之抗體可被獲得為嵌合抗體,介於來自 非人抗體之可變區與來自人類抗體之固定區之間,或為人 源化抗體,包括來自非人抗體之互補決定區、架構作用區 (frame work regi〇n’ FR)與來自人類抗體之固定區。根據 已知方法可製備此類抗體。藉由齧齒類互補決定區之序列 取代人類抗體對應之互補序列可執行人源化(參見,例如Trends Biotechnol 9: 132-7 (1991)). The antibody can be modified by binding to various molecules such as p〇ly ethylene glycol (PEG). The present invention provides such modified antibodies. A modified antibody can be obtained by chemically modifying an antibody. These methods of modification are common in the art. Alternatively, an antibody of the invention can be obtained as a chimeric antibody, between a variable region from a non-human antibody and a fixed region from a human antibody, or a humanized antibody, including a complementarity determining region from a non-human antibody. , framework work area (frame work regi〇n' FR) and fixed areas from human antibodies. Such antibodies can be prepared according to known methods. Humanization can be performed by substituting the sequence of the rodent complementarity determining region for the complementary sequence corresponding to the human antibody (see, for example,

Verhoeyen et al·, Science 239:1534-1536 (1988))。 因此,此類人源化抗體為嵌合抗體,其中實質上少於完整 之人類可變區已被以來自非人種類之對應序列取代。 也可使用包括除了架構作區與目定區尚有人類可變 73 201200525 區的全人類抗體。使用各種本技術領域所知的技術可產生 此類抗體。例如,i n v i t ro方法包括呈現於嗤菌體上之人 類抗體片段的重組資料庫的使用(例如,Hoogenboom &amp;Verhoeyen et al., Science 239: 1534-1536 (1988)). Thus, such humanized antibodies are chimeric antibodies in which substantially less than intact human variable regions have been substituted with corresponding sequences from non-human species. It is also possible to use fully human antibodies including the human variable 73 201200525 region in addition to the framework and the target region. Such antibodies can be produced using a variety of techniques known in the art. For example, the i n v i ro method includes the use of a recombinant database of human antibody fragments presented on bacillus (eg, Hoogenboom &amp;

Winter,J. Mol. Biol· 227:381 ( 1991 ))。相似地,藉 由將人類免疫球蛋白基因座(loci)引入轉殖動物,例如於 其中内生免疫球蛋白基因已被部分或完全去活化的小鼠, 可製造人類抗體。此方法被敘述,例如於U. S. Patent Nos. 6,1 50,584, 5, 545, 807; 5, 545, 806; 5, 569, 825; 5, 625, 126; 5, 633, 425; 5, 661,016。 獲自上述之抗體可被純化至同質(homogenei ty )。例 如’根據用於一般蛋白質的分離與純化方法可執行抗體之 分離與純化。例如,藉由合適地選擇與結合管柱色層分析, 例如親合管住、過濾、超過濾、鹽析、透析、對鈉十二烷 基的硫酸鹽聚丙稀醜胺凝膠電泳(SDS polyacrylamide gel electrophoresis)、等電焦集法(isoeiectric focusing) 的使用可分開與分離抗體(Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory ( 1 988)) ’但不限於此。蛋白質a管柱與蛋白 質G管柱A可被使用為親合管柱。要被使用之示範的蛋白 質 A 管柱包括,例如 Hyper D、P0R0S 與 Sepharose F. F. (Pharmacia)0 除了親合’示範之色層分析包括,例如離子交換色層 分析、疏水色層分析、膠體過濾、逆向色層分析、吸附色 層分析等(Strategies f0r protein Purificati〇n and 74 201200525Winter, J. Mol. Biol. 227:381 (1991)). Similarly, human antibodies can be made by introducing a human immunoglobulin locus (loci) into a transgenic animal, such as a mouse in which the endogenous immunoglobulin gene has been partially or completely deactivated. This method is described, for example, in US Patent Nos. 6, 1 50, 584, 5, 545, 807; 5, 545, 806; 5, 569, 825; 5, 625, 126; 5, 633, 425; 5, 661, 016. The antibodies obtained above can be purified to homogeneity. For example, isolation and purification of antibodies can be performed according to methods for isolation and purification of general proteins. For example, by suitable selection and combined column chromatography, such as affinity tube, filtration, ultrafiltration, salting out, dialysis, sodium lauryl sulfate polyacrylamide gel electrophoresis (SDS polyacrylamide) The use of gel electrophoresis and isoeiectric focusing can separate and isolate antibodies (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory (1 988)) 'but is not limited thereto. The protein a column and the protein G column A can be used as an affinity column. Exemplary protein A columns to be used include, for example, Hyper D, P0R0S and Sepharose FF (Pharmacia) 0 in addition to affinity 'exemplary chromatographic analysis including, for example, ion exchange chromatography, hydrophobic chromatography, colloid filtration, Reverse Chromatography, Adsorption Chromatography, etc. (Strategies f0r protein Purificati〇n and 74 201200525

Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratofy Press ( 1 996))。藉由液相色層分析,例如 與FPLC可執行色層分析步驟。 可使用吸收之測量、酵素連結免疫吸附分析 例如 (ELISA)、酵素免疫分析(EU)、放射免疫分析及/或免疫螢 光以測里本發明抗體之抗原結合活性。在酵素連結免疫吸 附分析中,本發明抗體為固定於一培養盤上,提供本發明 胜狀至培養盤,且之後提供含所需抗體之樣本,如,產生 抗體之細胞的培養懸浮液或經純化的抗體。之後,提供辨 第杬體且被標誌酵素,例如鹼性磷酸酶之第二抗體, 且之後培養培養盤。接著在清洗後’將酵素受質,例如對 硝基苯磷酸(p-ni tr〇pheny i ph〇sphate),加至培養盤,並 測量吸收以評估樣本之抗原結合活性。胜肽之片段,例如 C端或N端之片段可被使用為抗原以評估抗體結合活性。 根據本發明,可使用BIAcore (Pharmacia)來評估抗體的 活性。 上述方法允許本發明胜肽之偵測或測量,藉由露出本 發明抗體至假定含本發明胜肽之樣本並偵測或測量由抗體 與胜肽所形成之免疫複合物。 由於根據本發明之胜肽偵測或測量方法可專一偵測或 測量胜肽,方法在使用胜肽之各種實驗中為有用的。 χπ.載體與宿主細胞 本發明也提供載體與宿主細胞,於其中將編碼出本發 75 201200525 明胜肽之核苷酸引入。本發明之載體對維持本發明核替 酸’特別是DNA於宿主細胞中以表現本發明胜肽,或對於 用於基因治療來投予本發明本發明核苷酸為有效的。Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratofy Press (1996). The chromatographic analysis step can be performed by liquid chromatography, for example with FPLC. Absorption measurements, enzyme-linked immunosorbent assays such as (ELISA), enzyme immunoassay (EU), radioimmunoassay, and/or immunofluorescence can be used to measure the antigen binding activity of the antibodies of the invention. In an enzyme-linked immunosorbent assay, the antibody of the present invention is immobilized on a culture plate, providing the present invention to a culture plate, and then providing a sample containing the desired antibody, for example, a culture suspension of the antibody-producing cells or Purified antibody. Thereafter, a secondary antibody which recognizes the steroid and is labeled with an enzyme such as alkaline phosphatase is provided, and then the culture disk is cultured. Next, after washing, the enzyme substrate, for example, p-ni tr〇pheny i ph〇sphate, was added to the culture plate, and the absorption was measured to evaluate the antigen binding activity of the sample. Fragments of peptides, such as C-terminal or N-terminal fragments, can be used as antigens to assess antibody binding activity. According to the present invention, BIAcore (Pharmacia) can be used to evaluate the activity of an antibody. The above method allows the detection or measurement of the peptide of the present invention by exposing the antibody of the present invention to a sample presumed to contain the peptide of the present invention and detecting or measuring the immune complex formed by the antibody and the peptide. Since the peptide detection or measurement method according to the present invention can specifically detect or measure the peptide, the method is useful in various experiments using the peptide. Χπ. Vectors and Host Cells The present invention also provides vectors and host cells in which nucleotides encoding the phenoxypeptides of the present invention 75 201200525 are introduced. The vector of the present invention is effective for maintaining the nuclear acid of the present invention, particularly DNA in a host cell to express the peptide of the present invention, or for administering the nucleotide of the present invention for gene therapy.

當E. coli為宿主細胞且載體被放大且大量製造於E coli (例如 ’ JM109、DH5 alpha、HB101 或 XLlBlue)中時, 載體應具有要被放大於Ε· coli中之“ori”與篩選轉形 E · c 01 i之標諸基因(例如,藉由例如安比西林 (ampicillin)、四環黴素(tetracycline) ' 卡那黴素 (kanamycin) '氣黴素(chloramphenicol)或類似物之藥物 篩選之一抗藥基因)。例如,可使用M13_系列載體、pUc_ 系列载體、pBR322、pBluescript、pCR-Script 等。此外, pGEM-T ' pDIRECT與pT7也可被用來次複製與萃取cDNA與 上述載體。當載體被用來產生本發明蛋白質時,一表現载 體為特別有用。 例如’要被表現於E. co 1 i中之一表現載體應具有上 述特徵以被放大於E. coli中。當使用E. coli,例如 JM109、DH5 alpha、HB101 或 XLlBlue 為宿主細胞時,載 體應具有啟動子(promoter),例如lacZ啟動子(Ward et al., Nature 341: 544-6 (1989); FASEB J 6: 2422-7 (1 992))、araB 啟動子(Better et al.,Science 240: 1041-3 (1988))、T7啟動子或類似物,其可有效表現所需 基因於E. col i.中。基於那方面’可使用,例如ρΟΕχιχΗ (Pharmacia), &quot;QIAexpress system” (Qiagen)、pEGFP 與 pET(於此例子’宿主較佳為BL21,其表現T7 RNA聚合酶), ⑤ 76 201200525 取代上述載體。另外,載體也可含用於胜肽分泌之訊號序 列。一引導要被分泌之胜肽至 E. col i的胞膜間區 (periplasm)的示範之訊號序列為pelB訊號序列(Lei et al.,J Bacteriol 169: 4379 (1987))。將載體引入目標 宿主細胞的方式包括’例如,氣化鈣方法,與電穿孔 (electroporation)方法 ° 除了 E· co 1 i,例如來自哺乳動物之表現載體(例如 pcDNA3 (Invitrogen)與 pEGF-B〇S (Nucleic Acids Res 18(17): 5322 ( 1 990)),pEF,pCDM8)、來自昆蟲細胞之表 現載體(例如、&quot;Bac-to-BAC桿狀病毒表現系統 (baculovirus expression system)&quot; (GIBCO BRL)、 pBacPAK8 )、來自植物之表現載體(例如,pMH1、pMH2 )、 來自動物病毒之表現載體(例如、pHSV、pMV、pAdexLcw )、 來自反轉錄病毒之表現載體(例如,pZ i pneo )、來自酵母 菌之表現載體(例如’ ”Pichia Expression Kit&quot; (Invitrogen)、pNVll、SP-Q01 )與來自枯草桿菌(BaciUus subti 1 is)之表現載體(例如,ppL6〇8,ρκτΗ50 )可被用來 產生本發明之多胜肽。 為了在於動物細胞,例如CH0、C0S或NIH3T3細胞中 表現載體,載體應具有表現於此類細胞中所必須的啟動 子,例如 SV40 啟動子(MUlligan et al·, Nature 277: 1〇8 ( 1 979)) ' MMLV-LTR 啟動子、£F1 alpha 啟動子(Mizushima et al·’ Nucleic Acids Res 18: 5322 (1990)) 、 CMV 啟 動子等,與筛選轉形物的標誌基因(例如藉由藥物篩選(例 77 201200525 之抗藥基因)較佳。具有這 例如 pMAM、pDR2、pBK-RSV、 如’新黴素(neomycin)、G418 些特徵之已知載體的例子包括, pBK-CMV 、 p〇PRSV 與 p〇p13 。 呈現下列實施例以說明本發明與以協助熟悉此技藝人 士製造與使用本發明。實施例並不意味於在其他方面限制 本發明範圍任何方式中。 【實施例】 材料與方法 細胞株 T2(HLA-A2)、人類B淋巴母細胞株與c〇S7、非洲綠猴 腎細胞株為自ATCC所購得。 來自MYBL2之胜肽的候選物選擇 使用“NetMHC 3. 0”結合預測伺服器(binding prediction server) (http://www.cbs.dtu.dk/services/ NetMHC/) (Buus et al. (Tissue Antigens., 62:378-84, 2003 ),Nielsen eisA (Protein Sci.,1 2:1 007-1 7,2003, Bioinformatics,20(9):1 388-97, 2004))預測來自 MYBL2 之9員與10員胜肽,其結合至HLA-A* 0201分子。這些胜 肽係根據一標準固相合成方法由 Biosynthesis (Lewisville, TX)來合成且藉由逆相高效能液體層析 (reversed phase high performance liquid chromatography, HPLC)來純化。分別藉由分析型HLPC與 質譜分析確認這些胜肽之純度(&gt; 90 % )與身份 (identity)。 將胜肽溶解於二甲基亞砜 78 ⑤ 201200525 (dimethylsulf〇xide,DMS0)中以 20 mg/ml 且儲存於 _8〇 °C。 I n v i tro細胞毒殺性τ淋巴球誘導 使用來自單核白血球之樹突細胞做為抗原呈現細胞以 誘導抗表現於人類白血球組織抗原(HLA)上之胜肽的細胞 毒殺性T淋巴球反應。產生樹突細胞如別處所述 (Nakahara S et al., Cancer Res 2003 Jul 15, 63(14): 4112-8)。特別地,由 Fic〇H-Piaque (pharmacia)溶液將 分離自一正常自願者(HLA_A* 〇2〇1陽性)之周邊血液單核 細胞’藉由貼附至一塑膠組織培養盤(Bect〇n Dickins〇n) 來分離以豐富其如一單核白血球部分。將經豐富單核白血 球之族群培養在1 〇 〇〇 U/m 1之人類顆粒-巨嗤細胞群落刺激 因子(granulocyte-macrophage colony-stimulating factor, GM-CSF) (R&amp;D System)與 1 000 U/ml 之白細胞介 素(interleukin,IL)-4 (R&amp;D System)存在下於含 2%之 熱去活性自身取得血清(aut〇l〇g〇us serum,as)之AIM-V 培養基(Invitrogen)中。培養7天後,於aim-V培養基中, 於 3 &quot;g /ml 之万-2 微球蛋白(beta 2-microglobulin) 存在下以20 /zg/ml之各合成胜肽脈衝(pulsed)細胞激素 誘導之樹突細胞3小時於37〇c ^所產生之細胞顯示表現樹 突細胞相關分子’例如CD80、CD83、CD86與HLA Class 11 於其細胞表面(資料未顯示)。之後以χ_射線(2〇 Gy)將 這些胜肽脈衝之樹突細胞去活性且將其以1 : 2 〇之比例與 自身取得CD8陽性T細胞混合,而cd8陽性T細胞藉由以 79 201200525 CD8 Positive Isolation Kit (Dynal)正選擇獲得。這些 培養物a又置於48孔盤(c〇rning);各孔含丨&amp; χ 1 〇4胜肽 脈衝之樹突細胞、3 X 1〇5 CD8陽性Τ細胞與1〇 ng/ml之 IL-7 (R&amp;D System)於 0.5 ml 之 AIM-V/2% 自身取得血清培 養基中。三天之後,以IL_2(CHIR〇N)添加至培養物至終濃 度為20 IU/ml。於第7天與第14天更以自身取得胜肽脈 衝之樹突細胞進一步刺激T細胞。以與上述相同之方法每 次製備樹突細胞。於第21天,第三輪之胜肽刺激後,將細 胞毒殺性T淋巴球進行抗胜肽脈衝之T2細胞測試(Tanaka H et aL, Br J Cancer 200 1 Jan 5, 84(1): 94-9; Umano Y et al., Br J Cancer 2001 Apr 20, 84(8): 1052-7; Uchida N et al. , Clin Cancer Res 2004 Dec 15, 10(24): 8577-86; Suda T et aJ., Cancer Sci 2006 May, 97(5): 411-9; Watanabe T et aJ.t Cancer Sci 2005 Aug, 96(8): 498-506)。 細胞毒殺性T淋巴球擴張步驟 使用與由 Riddell et al. (Walter EA et a 1.,N Engl J Med 1 99 5 Oct 19,333(1 6): 1038-44; Riddell SR et al· ’When E. coli is a host cell and the vector is amplified and produced in large quantities in E coli (eg 'JM109, DH5 alpha, HB101 or XLlBlue), the vector should have the "ori" to be amplified in Ε·coli and screened The genes of the form E · c 01 i (for example, by drug screening such as ampicillin, tetracycline 'kanamycin ' chloramphenicol or the like One of the drug resistance genes). For example, an M13_series vector, a pUc_ series vector, pBR322, pBluescript, pCR-Script, or the like can be used. In addition, pGEM-T 'pDIRECT and pT7 can also be used for secondary replication and extraction of cDNA with the above vectors. A performance vector is particularly useful when the vector is used to produce a protein of the invention. For example, one of the expression vectors to be represented in E. co 1 i should have the above characteristics to be magnified in E. coli. When using E. coli, such as JM109, DH5 alpha, HB101 or XLlBlue as the host cell, the vector should have a promoter, such as the lacZ promoter (Ward et al., Nature 341: 544-6 (1989); FASEB J 6: 2422-7 (1 992)), araB promoter (Better et al., Science 240: 1041-3 (1988)), T7 promoter or analog, which can effectively express the desired gene in E. col i. Based on that aspect, 'can be used, for example, ρΟΕχιχΗ (Pharmacia), &quot;QIAexpress system&quot; (Qiagen), pEGFP and pET (in this example, the host is preferably BL21, which expresses T7 RNA polymerase), 5 76 201200525 replaces the above vector In addition, the vector may also contain a signal sequence for peptide secretion. An exemplary signal sequence that directs the peptide to be secreted to the periplasm of E. col i is the pelB signal sequence (Lei et al). J Bacteriol 169: 4379 (1987)). The manner in which the vector is introduced into the host cell of interest includes, for example, the calcium-calcification method, and the electroporation method, except for E·co 1 i, for example, from mammals. Vectors (eg, pcDNA3 (Invitrogen) and pEGF-B〇S (Nucleic Acids Res 18(17): 5322 (1 990)), pEF, pCDM8), expression vectors from insect cells (eg, &quot;Bac-to-BAC) Baculovirus expression system&quot; (GIBCO BRL), pBacPAK8), plant-derived expression vectors (eg, pMH1, pMH2), expression vectors from animal viruses (eg, pHSV, pMV, pAdexLc) w), a expression vector from a retrovirus (for example, pZ i pneo ), a expression vector derived from yeast (for example, ' 'Pichia Expression Kit&quot; (Invitrogen), pNVll, SP-Q01) and from Bacillus subtilis (BaciUus subti 1) A performance vector of is) (e.g., ppL6〇8, ρκτΗ50) can be used to produce the multi-peptide of the present invention. In order to express a vector in an animal cell, such as a CH0, COS or NIH3T3 cell, the vector should have a promoter necessary for expression in such a cell, such as the SV40 promoter (MUlligan et al., Nature 277: 1〇8 (1 979) )) 'MMLV-LTR promoter, £F1 alpha promoter (Mizushima et al·' Nucleic Acids Res 18: 5322 (1990)), CMV promoter, etc., and marker genes for screening transformants (eg by drug) Screening (drug resistance gene of Example 77 201200525) is preferred. Examples of known vectors having such features as pMAM, pDR2, pBK-RSV, such as 'neomycin, G418' include pBK-CMV, p〇 PRSV and p〇p13 The following examples are presented to illustrate the invention and are intended to be illustrative of the invention and the invention is not intended to limit the scope of the invention in any manner. Methods Cell line T2 (HLA-A2), human B lymphoblastoid cell line and c〇S7, African green monkey kidney cell line were purchased from ATCC. The candidate for peptide from MYBL2 was selected to use "NetMHC 3. 0". Combined predictive servo (binding prediction server) (http://www.cbs.dtu.dk/services/ NetMHC/) (Buus et al. (Tissue Antigens., 62:378-84, 2003), Nielsen eisA (Protein Sci., 1 2:1 007-1 7,2003, Bioinformatics, 20(9):1 388-97, 2004)) Predicting 9 and 10 member peptides from MYBL2, which bind to HLA-A* 0201 molecules. These wins Peptides were synthesized by Biosynthesis (Lewisville, TX) according to a standard solid phase synthesis method and purified by reversed phase high performance liquid chromatography (HPLC), respectively by analytical HLPC and mass spectrometry. Confirm the purity (&gt; 90%) and identity of these peptides. Dissolve the peptide in dimethyl sulfoxide 78 5 201200525 (dimethylsulf〇xide, DMS0) at 20 mg/ml and store at _8〇 °C. I n v i tro cytotoxic tau lymphocyte induction The dendritic cells derived from mononuclear leukocytes were used as antigen-presenting cells to induce cellular toxic T lymphocyte responses against peptides expressed on human leukocyte antigen (HLA). Dendritic cells are produced as described elsewhere (Nakahara S et al., Cancer Res 2003 Jul 15, 63(14): 4112-8). In particular, peripheral blood mononuclear cells isolated from a normal volunteer (HLA_A* 〇2〇1 positive) are attached by a Fic〇H-Piaque (Pharmacia) solution to a plastic tissue culture plate (Bect〇n) Dickins〇n) to separate to enrich it as a mononuclear white blood cell part. The population of abundant mononuclear white blood cells is cultured in 1 〇〇〇U/m 1 of human granulocyte-macrophage colony-stimulating factor (GM-CSF) (R&amp;D System) with 1 000 AIM-V medium in the presence of U/ml interleukin (IL)-4 (R&amp;D System) in serum containing 2% of heat deactivated itself (aut〇l〇g〇us serum, as) (Invitrogen). After 7 days of culture, the synthesized peptides were pulsed at 20 /zg/ml in the presence of 3 &quot;g /ml of 2-diglobulin in aim-V medium. Hormone-induced dendritic cells produced at 37 hC for 3 hours showed dendritic cell-associated molecules such as CD80, CD83, CD86 and HLA Class 11 on their cell surface (data not shown). The dendritic cells pulsed with these peptides were then deactivated with χ-rays (2〇Gy) and mixed with CD8-positive T cells in a ratio of 1:2 ,, while cd8-positive T cells were treated with 79 201200525 The CD8 Positive Isolation Kit (Dynal) is being selected. These cultures a were placed in a 48-well plate (c〇rning); each well contained 丨&amp; χ 1 〇 4 peptide-pulsed dendritic cells, 3 X 1〇5 CD8-positive sputum cells and 1 〇ng/ml IL-7 (R&amp;D System) was taken in serum medium at 0.5 ml of AIM-V/2% itself. Three days later, IL-2 (CHIR〇N) was added to the culture to a final concentration of 20 IU/ml. On day 7 and day 14, dendritic cells with peptide peptides were further stimulated by T cells. Dendritic cells were prepared each time in the same manner as above. On day 21, after the third round of peptide stimulation, the cytotoxic T lymphocytes were tested for anti-peptide pulsed T2 cells (Tanaka H et aL, Br J Cancer 200 1 Jan 5, 84(1): 94 -9; Umano Y et al., Br J Cancer 2001 Apr 20, 84(8): 1052-7; Uchida N et al., Clin Cancer Res 2004 Dec 15, 10(24): 8577-86; Suda T et aJ., Cancer Sci 2006 May, 97(5): 411-9; Watanabe T et aJ.t Cancer Sci 2005 Aug, 96(8): 498-506). The cytotoxic T lymphocyte expansion step is used by Riddell et al. (Walter EA et a 1., N Engl J Med 1 99 5 Oct 19, 333(1 6): 1038-44; Riddell SR et al·

Nat Med 1 996 Feb,2(2): 216-23)所敘述之相似方法於培 養中擴張細胞毒殺性T淋巴球。全部5 x 1 〇4細胞毒殺性 Τ淋巴球懸浮於25 m 1之含有兩種人類Β類淋巴母細胞株 之AIM-V/5%自身取得血清培養基,由MMC去活化,在40 ng/ml之抗-CD3單株抗體(Pharmingen)存在下。在開始培 養1天後,120 IU/ml之IL-2加入培養中。於第5、8、11 ⑤ 201200525 天以新鮮之含30 IU/ml之IL-2的AIM-V/5%自身取得血清 培養基提供給培養物(Tanaka H et al·,Br J Cancer 200 1 Jan 5, 84(1): 94-9; Umano Y et al., Br J Cancer 200 1 Apr 20, 84(8): 1052-7; Uchida N et al., Clin Cancer Res 2004 Dec 15, 10(24): 8577-86; Suda T et al. , Cancer Sci 2006 May, 97(5): 411-9; Watanabe T et al., Cancer Sci 2005 Aug, 96(8): 498-506)。 細胞毒殺性T淋巴球複製(clone)的建立 執行稀釋以具有0. 3、1與3細胞毒殺性T淋巴球/孔 於 96 圓底(round-bottomed)微效價盤(Nalge Nunc International)中。細胞毒殺性T淋巴球與1 χ 1〇4細胞/ 孔之2種兩種人類B類淋巴母細胞株、30 ng/m 1之抗-CD 抗體與125 U/ml之IL-2於全部150 // 1 /孔之含5%自身 取得之血清的AIM-V培養基中一起培養。10天後將50 // 1/ 孔之IL-2加入培養基中以達到125U/ml IL-2之終濃度。 於第14天測試細胞毒殺性T淋巴球之活性,且使用上述相 同方法擴張細胞毒殺性T淋巴球複製(Uchi da N e i a / ., Clin Cancer Res 2004 Dec 15, 10(24): 8577-86; Suda T et al., Cancer Sci 2006 May, 97(5): 411-9; Watanabe T ei a/., Cancer Sci 2005 Aug, 96(8): 498-506)。 專一之細胞毒殺性T淋巴球活性 為了測試專一之細胞毒殺性T淋巴球活性,執行 IFN-r酵素結合免疫斑點(ELISP0T)分析與IFN-γ酵素結 合免疫吸附(EL IS A)分析。特別地,製備胜肽脈衝之T2 (1 81 201200525 x l〇Vwell)為刺激細胞。培養之細胞於48孔中做為應答 細胞。在製造商步驟下執行IFN_ 7酵素結合免疫斑點分析 與IFN-7&quot;酵素結合免疫吸附分析。 強有力地表現目標基因與HLA_〇2任一或兩者的細胞的建 立 藉由PCR將編碼出目標基因之開放讀框或HLA_A&gt;|! 之cDNA放大。將PCR放大產物複製進一表現載體。使用 lipofectamine 2000 (invitr〇gen),根據製造商建議步驟 將質體轉染進C0S7,其為一目標基因與HLA_A* 〇2〇1無效 細胞株。於自轉染後2天,以versene (Invi trogen)收集 經轉染的細胞且使用為細胞毒殺性T淋巴球活性分析之刺 激細胞(5 X 1 〇4/we 11)。 結果 來自MYBL2之HLA-A02結合胜肽的預測 表la與lb以高之結合親和力之順序顯示MYBL2之 HLA-A02結合9員與10員胜狀。總共4個具有潛在JJLA-A02 結合能力之胜肽被選擇且將其試驗以確定抗原決定位胜 肽。 201200525 表la、來自MYBL2之HLA-A02結合Θ員胜肽 起始位置 胺基酸序列 Kd (nM) 序列辨識號 355 VLPPRQPSA 213 1 597 LMMSTLPKS 285 2 144 RIICEAHKV 493 3 表lb、來自MYBL2之HLA-A02結合10員胜肽 起始位置 胺基酸序列 Kd (nM) 序列辨識號 596 KLMMSTLPKS 226 4 起始位置指自MYBL2之N端的胺基酸殘基數目。結合 分數與分離常數(dissociation constant)[Kd (nM)]來自 “BIMAS” 與 “NetMHC3.0” 。 以HLA- A* 0201限制之來自MYBL2之預測胜肽誘導細胞毒 殺性T淋巴球 根據敘述於“材料與方法”之步驟產生對於那些來自 MYBL2之胜肽的細胞毒殺性τ淋巴球。藉由iFN_r酵素結 σ免疫斑點分析測定胜肽專一細胞毒殺性τ淋巴球活性 (第1圖)。與控制組孔洞相較,以MYBL2-A02-9-144 (序 列辨識號:3 )刺激之孔洞編號#8 (a)顯示強而有力的 IFN-γ產生。另一方面,藉由以顯示於表1中之其他胜肽 刺激沒有測定出專一之細胞毒殺性T淋巴球活性,儘管那 些胜肽具有與HLA-A* 〇201之可能結合活性。如負資料的典 型例子,從以MYBL2-A02-9-355 (序列辨識號:1)(⑴刺激 之細胞毒殺性T淋巴球,並無觀察到專一的IFN_ 7產生。 因此,其指出篩選出來自MYBL2之MYBL2-A02-9-144 (序 列辨識唬· 3)為可誘導強而有力之細胞毒殺性τ淋巴球的 胜肽。 83 201200525 抗MYBL2特定胜肽之細胞毒殺性T淋巴球細胞株與複製的 建立 藉由IFN- 7 酵素結合免疫斑點分析偵測,於以 MYBL2-A0 2-9-144 (序列辨識號:3)刺激之孔洞編號#8中 顯示胜肽專一細胞毒殺性Τ淋巴球活性的細胞被擴張,並 藉由擴張程序來建立細胞毒殺性τ淋巴球細胞株,於上方 段落“材料與方法”中所述。藉由IFN-r酵素結合免疫吸 附分析來測定那些細胞毒殺性τ淋巴球細胞株的細胞毒殺 性T淋巴球活性(第2圖)。與無胜肽脈衝之目標細胞相 較’細胞毒殺性τ淋巴球細胞株顯示強的抗以對應胜肽脈 衝之目標細胞的IFN- 7產生。此外,如於“材料與方法,’ 所述,藉由來自細胞毒殺性T淋巴球細胞株的限制稀釋建 立細胞毒殺性T淋巴球複製,且藉由iFN_ r酵素結合免疫 吸附分析測定來自抗經胜肽脈衝目標細胞之細胞毒殺性τ 淋巴球複製的IFN_r產生。從以MYBL2-A02-9-144 (序列 辨識號.3 )刺激之細胞毒殺性τ淋巴球複製測定出強的 IFN- r產生(第3圖)。 抗外生表現MYBL2與HLA-A* 0201之目標細胞的專一細胞毒 殺性T淋巴球活性 檢驗經提升抗MYBL2-A02-9-144 (序列辨識號:3)之 所建立的細胞毒殺性T淋巴球細胞株其對於辨認表現 MYBL2與HLA-A* 0201分子之目標細胞的能力。使用由 MYBL2-A02-9-144 (序列辨識號:3)提升的細胞毒殺性τ 淋巴球細胞株複製為應答細胞來測試抗C〇s7細胞的專一 84 201200525 細胞毒殺性T淋巴球活性,而c〇S7細胞經全長之MYBL2與 HLAH201分子基因兩者轉染(對於表現mYBL2與HLA-A * 020 1基因之目標細胞的特定模式)。c〇S7細胞以全長 MYBL2或HLA-A* 020 1轉染製備為控制組。於第4圖中,以A similar method as described in Nat Med 1 996 Feb, 2(2): 216-23) expands cytotoxic T lymphocytes in culture. All 5 x 1 〇4 cytotoxic sputum lymphocytes were suspended in 25 m 1 of AIM-V/5% of two human sputum lymphoblastoid cell lines to obtain serum medium, deactivated by MMC at 40 ng/ml In the presence of anti-CD3 monoclonal antibody (Pharmingen). One day after the start of cultivation, 120 IU/ml of IL-2 was added to the culture. On the 5th, 8th, and 11th, 5th, 5th, 2012th, 2012, the serum medium was obtained from the fresh AIM-V/5% self containing 30 IU/ml of IL-2 (Tanaka H et al., Br J Cancer 200 1 Jan). 5, 84(1): 94-9; Umano Y et al., Br J Cancer 200 1 Apr 20, 84(8): 1052-7; Uchida N et al., Clin Cancer Res 2004 Dec 15, 10(24 ): 8577-86; Suda T et al., Cancer Sci 2006 May, 97(5): 411-9; Watanabe T et al., Cancer Sci 2005 Aug, 96(8): 498-506). Establishment of cytotoxic T lymphocyte replication was performed to have a 0.3, 1 and 3 cytotoxic T lymphocyte/well in a 96 round-bottomed micro-valence plate (Nalge Nunc International) . Cytotoxic T lymphocytes and 1 χ 1〇4 cells/well of two human B-type lymphoblastoid cells, 30 ng/m 1 anti-CD antibody and 125 U/ml IL-2 in all 150 // 1 / well of AIM-V medium containing 5% of the serum obtained by itself. After 10 days, 50 // 1/well of IL-2 was added to the medium to reach a final concentration of 125 U/ml IL-2. The activity of the cytotoxic T lymphocytes was tested on day 14 and the cytotoxic T lymphocyte replication was expanded using the same method described above (Uchi da N eia / ., Clin Cancer Res 2004 Dec 15, 10(24): 8577-86 Suda T et al., Cancer Sci 2006 May, 97(5): 411-9; Watanabe T ei a., Cancer Sci 2005 Aug, 96(8): 498-506). Specific Cytotoxic T Lymphocyte Activity To test the specific cytotoxic T lymphocyte activity, IFN-r enzyme binding immunospot (ELISP0T) analysis and IFN-γ enzyme binding immunosorbent (EL IS A) analysis were performed. Specifically, T2 (1 81 201200525 x l〇Vwell) of the peptide pulse was prepared to stimulate the cells. The cultured cells were used as responder cells in 48 wells. IFN-7 enzyme binding immunospot analysis was performed under the manufacturer's step with IFN-7&quot; enzyme binding immunosorbent assay. Establishment of cells that strongly express either or both of the target gene and HLA_〇2 The cDNA encoding the open reading frame of the target gene or HLA_A&gt;|! is amplified by PCR. The PCR amplification product is copied into a performance vector. Using lipofectamine 2000 (invitr〇gen), the plastid was transfected into COS7 according to the manufacturer's recommended procedure, which is a target gene and HLA_A* 〇2〇1 null cell line. Two days after transfection, transfected cells were harvested as versene (Invi trogen) and stimulator cells (5 X 1 〇 4/we 11) analyzed for cytotoxic T lymphocyte activity were used. Results Prediction of HLA-A02 Binding Peptides from MYBL2 Tables la and lb show that HLA-A02 of MYBL2 binds 9 members and 10 members in order of high binding affinity. A total of 4 peptides with potential JJLA-A02 binding ability were selected and tested to determine epitopes. 201200525 Table la, HLA-A02 from MYBL2 binding partner peptide amino acid sequence Kd (nM) Sequence ID 355 VLPPRQPSA 213 1 597 LMMSTLPKS 285 2 144 RIICEAHKV 493 3 Table lb, HLA-A02 from MYBL2 Binding of the 10-member peptide start position amino acid sequence Kd (nM) Sequence ID 596 KLMMSTLPKS 226 4 The starting position refers to the number of amino acid residues from the N-terminus of MYBL2. The binding fraction and the dissociation constant [Kd (nM)] are derived from "BIMAS" and "NetMHC3.0". The predicted peptide derived from MYBL2, restricted by HLA-A* 0201, induces cytotoxic T lymphocytes. The cytotoxic tau lymphocytes from those peptides derived from MYBL2 are produced according to the procedure described in "Materials and Methods". The peptide-specific cytotoxic lymphosphere activity was determined by iFN_r enzyme sigma immunospot assay (Fig. 1). Compared with the control group, the hole number #8 (a) stimulated by MYBL2-A02-9-144 (serial identification number: 3) showed strong IFN-γ production. On the other hand, specific cytotoxic T lymphocyte activity was not determined by stimulation with other peptides shown in Table 1, although those peptides had a possible binding activity to HLA-A* 〇201. As a typical example of negative data, from cytotoxic T lymphocytes stimulated with MYBL2-A02-9-355 (SEQ ID NO: 1) ((1), no specific IFN-7 was observed. Therefore, it was indicated that MYBL2-A02-9-144 from MYBL2 (sequence recognition 唬·3) is a peptide that induces potent and potent cytotoxic lysate. 83 201200525 Cytotoxic T lymphocyte cell line against MYBL2 specific peptide The establishment of the replication was detected by IFN-7 enzyme-binding immunospot assay, and the peptide-specific cytotoxic sputum was displayed in the hole number #8 stimulated by MYBL2-A0 2-9-144 (SEQ ID NO: 3). The spheroidally active cells are expanded and the cytotoxic TNF lymphocyte cell line is established by expansion procedures as described in the above section "Materials and Methods." Those cytotoxic kills are determined by IFN-r enzyme binding immunosorbent assay. Cytotoxic T lymphocyte activity of the tau lymphocyte cell line (Fig. 2). Compared with the target cell of the peptide-free pulse, the 'cytotoxic tau lymphocyte cell line showed a strong resistance to the target of the corresponding peptide pulse. Cell IFN-7 production. This , as described in "Materials and Methods," by establishing a cytotoxic T lymphocyte replication by limiting dilution from a cytotoxic T lymphocyte cell line, and determined by iFN_r enzyme binding immunosorbent assay IFN-r production by cytotoxic τ lymphocyte replication of peptide-pulsed target cells. Strong IFN- r production was determined from cytotoxic tibia lymphocyte replication stimulated with MYBL2-A02-9-144 (SEQ ID NO: 3) Figure 3). Specific anti-exogenous MYBL2 and HLA-A* 0201 target cells were tested for specific cytotoxic T lymphocyte activity by increasing anti-MYBL2-A02-9-144 (SEQ ID NO: 3) The ability of the cytotoxic T lymphocyte cell line to recognize target cells expressing MYBL2 and HLA-A* 0201 molecules. The cytotoxic τ lymphocytes promoted by MYBL2-A02-9-144 (SEQ ID NO: 3) were used. The cell line was replicated as a responsive cell to test the specific 84 201200525 cytotoxic T lymphocyte activity of anti-C〇s7 cells, while the c〇S7 cells were transfected with both the full-length MYBL2 and HLAH201 molecular genes (for the expression of mYBL2 and HLA-A). * 020 1 gene target Cell specific mode) MYBL2 .c〇S7 cells or the full-length HLA-A * 020 1 transfected prepared as a control group. In FIG. 4, to

MYBL2-A02-9-144 (序列辨識號:3 )刺激之細胞毒殺性T 淋巴球複製顯示強的抗表現MYBL2與HLA-A* 020 1兩者之 COS7細胞的細胞毒殺性τ淋巴球活性。另一方面,沒有偵 測到抗控制組之顯著專一之細胞毒殺性T淋巴球活性。因 此,這些資料清楚證明MYBL2-A02-9-144 (序列辨識號:3) 被自然表現於具有HLA-A* 020 1分子之目標細胞上且由細 胞毒殺性T淋巴球所辨認。這些結果顯示來自MYBL2之 MYBL2-A02-9-144 (序列辨識號:3)為可適合為具MYBL2 表現之腫瘤之病患之治療的癌症疫苗。 抗原胜肽之同源分析 以MYBL2-A02-9-144 (序列辨識號:3)刺激之細胞毒 殺性T淋巴球顯示顯著且專一之細胞毒殺性τ淋巴球活 性。此結果可能起因於MYBL2-A02-9-144 (序列辨識號:3) 之序列為與源自已知使人類免疫系統敏感之其他分子的胜 肽同源的事實。為了排除此可能性,對於使用為關鍵字向 BLAST 演算法(http://www.ncbi.nlm.nih.gov/blast/ blast, cgi)查詢之此胜肽序列執行同源性分析,而BLAST 演算法顯示沒有序列顯示顯著之同源性。同源性分析之結 果指出MYBL2-A02-9-144 (序列辨識號:3)之序列為獨特 的’且因此對於我們最大知識而言只有很小可能性分子會 85 201200525 對於一些非相關分子提高不想要之免疫反應。 因此’確認MYBL2-A02-9-144 (序列辨識號:3)為新 穎之來自MYBL2之HLA-A* 0201抗原決定位胜肽。此處結果 證明MYBL2-A02-9-144 (序列辨識號:3)對使用於癌症免 疫治療中而言可為適合的。 產業利用性 本發明提供新的腫瘤相關抗原’特別是來自MYBL2的 那些,其可誘導強且專一的抗腫瘤免疫反應,且對於癌症 形式之廣泛種類而言具有應用性。此腫瘤相關抗原更成為 發展抗與MYBL2相關之疾病的胜肽疫苗的根據,與MYBL2 相關之疾病,例如癌症,更特別是睪丸腫瘤、胰臟癌、膀 胱癌、非小細胞肺癌、小細胞肺癌與食道癌。 當於此詳述本發明與提及其特定實施例時,需瞭解的 是,前述敘述本質為示範與解釋且意為說明本發明與其較 佳實施例。於例行實驗之中,熟悉此技藝人士可立即瞭解 在不脫離本發明之精神下其可實行各種改變與修飾。因此 本發明不被前述所定義,而被下列t請專利範圍與其等同 物所定義。 【圖式簡單說明】 第1圖由-系列照片’⑷至⑻所組成,其顯示在以 來自MYBL2之胜肽誘導之細胞毒殺性τ淋巴球上之IFN一y 酵素結合免疫斑點分析(ELISP0T)的結果《與控制組相較, 86 ⑤ 201200525 於孔洞編號#8中以MYBL2-A〇2_9_144 (序列辨識號:3)刺 激之細胞毒殺性τ淋巴球&amp;)顯示強的IFN—γ產生。於這 些圖片之孔洞上之正方形指出於來自對應孔洞之細胞被擴 張來建立細胞毒殺性Τ淋巴球細胞株。相對的,如負資料 的典型例子,從以Mm2-AQ2_9_355 (序列辨識號:1)刺 激之細胞毒殺性τ淋巴球沒有偵測到專一的IFN_T產 生。於®巾,+指出抗以適合之胜肽脈衝的目標細胞的 IF N - 7 產生,而“”指山4二A # 夺曰出抗未以任何胜肽脈衝的目標細 胞的IFN-γ產生。 第2圖為一線圖,顯示IFN_T酵素結合免疫吸附分析 债測的結果,其顯示以MYBL2_a〇2_9_U4 (序列辨識號:3 ) 刺激之細胞毒殺性τ淋巴球細胞株的IFN_r產生。結果顯 不與控制組相較’藉由以此胜肽刺激建立之細胞毒殺性T 淋巴球細胞株顯示強而有力之IFN_r產生。於圖中,‘‘+,, 指出抗以適合之胜肽脈衝的目標細胞的IFN_y產生,而 -指出抗未以任何胜肽脈衝的目標細胞的產 生0 第3圖為一線圖’顯示藉由來自以MYBL2-A02-9-144 (序列辨識號:3 )刺激之細胞毒殺性τ淋巴球細胞株的限 制稀釋所建立之細胞毒殺性τ淋巴球複製的IFN- r產生。 、、’。果顯不與控制組相較,藉由以此胜肽刺激所建立之細胞 毋殺性T淋巴球複製顯示強而有力之IFN-γ產生。於圖 中 5 “ +,, 士匕 ^出抗以適合之胜肽脈衝的目標細胞的丨FN_ ^ 產生 而 ~指出抗未以任何胜肽脈衝的目標細胞的 87 201200525 IFN- 7產生β 第4圖為一線圖,其顯示抗表現MYBL2與HLA-A* 0201 之目標細胞的專一細胞毒殺性T淋巴球活性。將以hlA-A* 0201或全長之MYBL2基因轉染之C0S7細胞製備為控制組。 以MYBL2-A02-9-144 (序列辨識號:3)建立之細胞毒殺性 T淋巴球細胞複製顯示抗以MYBL2與HLA-A* 0201兩者轉染 之C0S7細胞的專一細胞毒殺性τ淋巴球活性(黑色菱形)。 另一方面,對於表現HLA-A* 0201 (三角形)或〇儿2 (圓 形)之任-的目標細胞,沒有偵測到顯著專一之細胞毒殺 性T淋巴球活性。 【主要元件符號說明】 無。 88 ⑤ 201200525 序列表 〈110〉 腫瘤療法·科學股份有限公司 &lt;120&gt; MYBL2胜肽及含此胜肽之疫苗 &lt;130〉 0NC-A0922-TW 〈150〉 US 61/266,871 &lt;151〉 2009-12-04 &lt;160〉 10 〈170〉 Patent In version 3.5 &lt;210〉 1 &lt;211&gt; 9 〈212〉 PRT 〈213〉 人工 &lt;220〉 〈223〉 人工合成胜肽序列 〈400〉 1MYBL2-A02-9-144 (SEQ ID NO: 3) stimulated cytotoxic T lymphocyte replication showed strong cytotoxic thymocyte activity in COS7 cells expressing both MYBL2 and HLA-A*020 1 . On the other hand, significant specific cytotoxic T lymphocyte activity in the anti-control group was not detected. Therefore, these data clearly demonstrate that MYBL2-A02-9-144 (SEQ ID NO: 3) is naturally expressed on target cells with HLA-A* 020 1 molecule and is recognized by cytotoxic T lymphocytes. These results show that MYBL2-A02-9-144 (SEQ ID NO: 3) from MYBL2 is a cancer vaccine that can be suitable for the treatment of patients with tumors with MYBL2 expression. Homologous analysis of antigenic peptides The cytotoxic T lymphocytes stimulated with MYBL2-A02-9-144 (SEQ ID NO: 3) showed significant and specific cytotoxic thymocyte activity. This result may be due to the fact that the sequence of MYBL2-A02-9-144 (SEQ ID NO: 3) is homologous to a peptide derived from other molecules known to be sensitive to the human immune system. To rule out this possibility, homology analysis was performed on this peptide sequence that was searched for the BLAST algorithm (http://www.ncbi.nlm.nih.gov/blast/blast, cgi) for the keyword, while BLAST The algorithm shows that no sequences show significant homology. The results of the homology analysis indicated that the sequence of MYBL2-A02-9-144 (SEQ ID NO: 3) is unique 'and therefore only a small probability for our greatest knowledge. The molecule will be 85 201200525 for some non-related molecules Unwanted immune response. Therefore, it was confirmed that MYBL2-A02-9-144 (SEQ ID NO: 3) was a novel HLA-A* 0201 epitope peptide derived from MYBL2. The results here demonstrate that MYBL2-A02-9-144 (SEQ ID NO: 3) may be suitable for use in cancer immunotherapy. Industrial Applicability The present invention provides novel tumor-associated antigens, particularly those from MYBL2, which induce a strong and specific anti-tumor immune response and are useful for a wide variety of cancer forms. This tumor-associated antigen is the basis for the development of a peptide vaccine against MYBL2-related diseases, diseases associated with MYBL2, such as cancer, more particularly, testicular tumors, pancreatic cancer, bladder cancer, non-small cell lung cancer, small cell lung cancer. With esophageal cancer. While the invention has been described with respect to the specific embodiments thereof, it will be understood that In the course of routine experimentation, it will be readily apparent to those skilled in the art that various changes and modifications can be made without departing from the spirit of the invention. Therefore, the present invention is not defined by the foregoing, but is defined by the following claims and their equivalents. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 consists of a series of photographs '(4) to (8) showing IFN-y-enzyme-binding immunospot assay (ELISP0T) on a cytotoxic lytic lymphocyte induced by a peptide derived from MYBL2. As a result, compared with the control group, 86 5 201200525 cytotoxic τ lymphocytes & stimulating by MYBL2-A〇2_9_144 (SEQ ID NO: 3) in hole number #8 showed strong IFN-γ production. The squares on the holes of these pictures indicate that the cells from the corresponding holes are expanded to establish a cytotoxic lymphocyte cell line. In contrast, as a typical example of negative data, no specific IFN_T was detected from the cytotoxic tau lymphocytes stimulated with Mm2-AQ2_9_355 (SEQ ID NO: 1). In the ® towel, + indicates that IF N - 7 is produced against the target cell pulsed with the appropriate peptide, while "" refers to the mountain 4 A A # 曰 曰 IFN-γ production against the target cells not pulsed with any peptide . Fig. 2 is a one-line diagram showing the results of IFN_T enzyme-binding immunosorbent assay debt assay showing IFN_r production by cytotoxic T cell lymphocytes stimulated with MYBL2_a〇2_9_U4 (SEQ ID NO: 3). The results were not as compared with the control group. The cytotoxic T lymphocyte cell line established by this peptide stimulation showed strong IFN_r production. In the figure, ''+,, indicates the production of IFN_y against target cells pulsed with a suitable peptide, and - indicates the production of target cells that are not pulsed with any peptide. Figure 3 is a one-line diagram' display by IFN-r production from cytotoxic tau lymphocytes replicated by limiting dilution of cytotoxic tau lymphocyte cell lines stimulated with MYBL2-A02-9-144 (SEQ ID NO: 3). ,,’. It was shown that the cell-killing T lymphocyte replication established by this peptide stimulation showed strong IFN-γ production compared to the control group. In the figure, 5 "+,, 匕 匕 ^ 抗 以 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 适合 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 87 The figure is a one-line diagram showing the specific cytotoxic T lymphocyte activity against target cells expressing MYBL2 and HLA-A* 0201. The C0S7 cells transfected with hlA-A* 0201 or the full-length MYBL2 gene were prepared as control groups. The cytotoxic T lymphocyte replication established by MYBL2-A02-9-144 (SEQ ID NO: 3) showed specific cytotoxic τ lymphocytes against C0S7 cells transfected with both MYBL2 and HLA-A* 0201 Ball activity (black diamond). On the other hand, no significant specific cytotoxic T lymphocyte activity was detected in target cells expressing HLA-A* 0201 (triangle) or deaf 2 (circle). [Major component symbol description] None. 88 5 201200525 Sequence Listing <110> Tumor Therapy Science Co., Ltd. &lt;120&gt; MYBL2 peptide and vaccine containing the peptide &lt;130〉 0NC-A0922-TW <150> US 61/266,871 &lt;151> 2009-12-04 &lt;160〉 10 <170> Patent In version 3.5 &lt;210> 1 &lt;211&gt; 9 <212> PRT <213> Labor &lt;220> <223> Synthetic peptide sequence <400> 1

Val Leu Pro Pro Arg Gin Pro Ser Ala 1 5 &lt;210〉 2 &lt;211〉 9 &lt;212&gt; PRT &lt;213〉人工 〈220〉 〈223&gt;人工合成胜肽序列 &lt;400〉 2Val Leu Pro Pro Arg Gin Pro Ser Ala 1 5 &lt;210〉 2 &lt;211> 9 &lt;212&gt; PRT &lt;213>Artificial <220> <223&gt; Synthetic peptide sequence &lt;400〉 2

Leu Met Met Ser Thr Leu Pro Lys Ser 1 5 &lt;210〉 3 201200525 &lt;211〉 9 &lt;212〉 PRT &lt;213〉人工 &lt;220〉 &lt;223〉人工合成胜肽序列 &lt;400〉 3Leu Met Met Ser Thr Leu Pro Lys Ser 1 5 &lt;210> 3 201200525 &lt;211> 9 &lt;212> PRT &lt;213>Manual &lt;220> &lt;223> Synthetic peptide sequence &lt;400> 3

Arg lie lie Cys Glu Ala His Lys Val 1 5 10PRT人 &lt;210&gt; &lt;211〉 &lt;212〉 &lt;213〉 &lt;220〉 &lt;223〉人工合成胜肽序列 &lt;400〉 4Arg lie lie Cys Glu Ala His Lys Val 1 5 10PRT person &lt;210&gt;&lt;211&gt;212>&lt;213>&lt;220>&lt;223>synthetic peptide sequence &lt;400> 4

Lys Leu Met Met Ser Thr Leu Pro Lys Ser 1 5 10 &lt;210〉 5 &lt;211&gt; 2731 &lt;212&gt; DNA &lt;213〉 人類 &lt;400&gt; 5 gcgcttggcg ggagatagaa aagtgcttca acccgcgccg gcggcgactg cagttcctgc 60 gagcgaggag cgcgggacct gctgacacgc tgacgccttc gagcgcggcc cggggcccgg 120 agcggccgga gcagcccggg tcctgacccc ggcccggctc ccgctccggg ctctgccggc 180 gggcgggcga gcgcggcgcg gtccgggccg gggggatgtc tcggcggacg cgctgcgagg 240 atctggatga gctgcactac caggacacag attcagatgt gccggagcag agggatagca 300 agtgcaaggt caaatggacc catgaggagg acgagcagct gagggccctg gtgaggcagt 360 2 ⑤ 201200525 ttggacagca ggactggaag ttcctggcca gccacttccc taaccgcact gaccagcaat 420 gccagtacag gtggctgaga gttttgaatc cagaccttgt caaggggcca tggaccaaag 480 aggaagacca aaaagtcatc gagctggtta agaagtatgg cacaaagcag tggacactga 540 ttgccaagca cctgaagggc cggctgggga agcagtgccg tgaacgctgg cacaaccacc 600 tcaaccctga ggtgaagaag tcttgctgga ccgaggagga ggaccgcatc atctgcgagg 660 cccacaaggt gctgggcaac cgctgggccg agatcgccaa gatgttgcca gggaggacag 720 acaatgctgt gaagaatcac tggaactcta ccatcaaaag gaaggtggac acaggaggct 780 tcttgagcga gtccaaagac tgcaagcccc cagtgtactt gctgctggag ctcgaggaca 840 aggacggcct ccagagtgcc cagcccacgg aaggccaggg aagtcttctg accaactggc 900 cctccgtccc tcctaccata aaggaggagg aaaacagtga ggaggaactt gcagcagcca 960 ccacatcgaa ggaacaggag cccatcggta cagatctgga cgcagtgcga acaccagagc 1020 ccttggagga attcccgaag cgtgaggacc aggaaggctc cccaccagaa acgagcctgc 1080 cttacaagtg ggtggtggag gcagctaacc tcctcatccc cgctgtgggt tctagcctct 1140 ctgaagccct ggacttgatc gagtcggacc ctgatgcttg gtgtgacctg agtaaatttg 1200 acctccctga ggaaccatct gcagaggaca gtatcaacaa cagcctagtg cagctgcaag 1260 cgtcacatca gcagcaagtc ctgccacccc gccagccttc cgccctggtg cccagtgtga 1320 ccgagtaccg cctggatggc cacaccatct cagacctgag ccggagcagc cggggcgagc 1380 tgatccccat ctcccccagc actgaagtcg ggggctctgg cattggcaca ccgccctctg 1440 tgctcaagcg gcagaggaag aggcgtgtgg ctctgtcccc tgtcactgag aatagcacca 1500 gtctgtcctt cctggattcc tgtaacagcc tcacgcccaa gagcacacct gttaagaccc 1560 tgcccttctc gccctcccag tttctgaact tctggaacaa acaggacaca ttggagctgg 1620 agagcccctc gctgacatcc accccagtgt gcagccagaa ggtggtggtc accacaccac 1680 201200525 tgcaccggga caagacaccc ctgcaccaga aacatgctgc gtttgtaacc ccagatcaga 1740 agtactccat ggacaacact ccccacacgc caaccccgtt caagaacgcc ctggagaagt 1800 acggacccct gaagcccctg ccacagaccc cgcacctgga ggaggacttg aaggaggtgc 1860 tgcgttctga ggctggcatc gaactcatca tcgaggacga catcaggccc gagaagcaga 1920 agaggaagcc tgggctgcgg cggagcccca tcaagaaagt ccggaagtct ctggctcttg 1980 acattgtgga tgaggatgtg aagctgatga tgtccacact gcccaagtct ctatccttgc 2040 cgacaactgc cccttcaaac tcttccagcc tcaccctgtc aggtatcaaa gaagacaaca 2100 gcttgctcaa ccagggcttc ttgcaggcca agcccgagaa ggcagcagtg gcccagaagc 2160 cccgaagcca cttcacgaca cctgccccta tgtccagtgc ctggaagacg gtggcctgcg 2220 gggggaccag ggaccagctt ttcatgcagg agaaagcccg gcagctcctg ggccgcctga 2280 agcccagcca cacatctcgg accctcatct tgtcctgagg tgttgagggt gtcacgagcc 2340 cattctcatg tttacagggg ttgtgggggc agagggggtc tgtgaatctg agagtcattc 2400 aggtgacctc ctgcagggag ccttctgcca ccagcccctc cccagactct caggtggagg 2460 caacagggcc atgtgctgcc ctgttgccga gcccagctgt gggcggctcc tggtgctaac 2520 aacaaagttc cacttccagg tctgcctggt tccctcccca aggccacagg gagctccgtc 2580 agcttctccc aagcccacgt caggcctggc ctcatctcag accctgctta ggatggggga 2640 tgtggccagg ggtgctcctg tgctcaccct ctcttggtgc atttttttgg aagaataaaa 2700 ttgcctctct cttaaaaaaa aaaaaaaaaa a 2731 &lt;210〉 6 &lt;211〉 700 &lt;212〉 PRT &lt;213〉人類 &lt;400&gt; 6 4 ⑤ 201200525Lys Leu Met Met Ser Thr Leu Pro Lys Ser 1 5 10 &lt;210> 5 &lt;211&gt; 2731 &lt;212&gt; DNA &lt;213&gt; Human &lt;400&gt; 5 gcgcttggcg ggagatagaa aagtgcttca acccgcgccg gcggcgactg cagttcctgc 60 gagcgaggag cgcgggacct gctgacacgc tgacgccttc gagcgcggcc cggggcccgg 120 agcggccgga gcagcccggg tcctgacccc ggcccggctc ccgctccggg ctctgccggc 180 gggcgggcga gcgcggcgcg gtccgggccg gggggatgtc tcggcggacg cgctgcgagg 240 atctggatga gctgcactac caggacacag attcagatgt gccggagcag agggatagca 300 agtgcaaggt caaatggacc catgaggagg acgagcagct gagggccctg gtgaggcagt 360 2 ⑤ 201200525 ttggacagca ggactggaag ttcctggcca gccacttccc taaccgcact gaccagcaat 420 gccagtacag gtggctgaga gttttgaatc cagaccttgt caaggggcca tggaccaaag 480 aggaagacca aaaagtcatc gagctggtta Agaagtatgg cacaaagcag tggacactga 540 ttgccaagca cctgaagggc cggctgggga agcagtgccg tgaacgctgg cacaaccacc 600 tcaaccctga ggtgaagaag tcttgctgga ccgaggagga ggaccgcatc atctgcgagg 660 cccacaaggt gctgggcaac cgctgggccg agatcgccaa gatgttgcca gggaggacag 720 acaatgctgt gaaga atcac tggaactcta ccatcaaaag gaaggtggac acaggaggct 780 tcttgagcga gtccaaagac tgcaagcccc cagtgtactt gctgctggag ctcgaggaca 840 aggacggcct ccagagtgcc cagcccacgg aaggccaggg aagtcttctg accaactggc 900 cctccgtccc tcctaccata aaggaggagg aaaacagtga ggaggaactt gcagcagcca 960 ccacatcgaa ggaacaggag cccatcggta cagatctgga cgcagtgcga acaccagagc 1020 ccttggagga attcccgaag cgtgaggacc aggaaggctc cccaccagaa acgagcctgc 1080 cttacaagtg ggtggtggag gcagctaacc tcctcatccc cgctgtgggt tctagcctct 1140 ctgaagccct ggacttgatc gagtcggacc ctgatgcttg gtgtgacctg agtaaatttg 1200 acctccctga ggaaccatct gcagaggaca gtatcaacaa cagcctagtg cagctgcaag 1260 cgtcacatca gcagcaagtc ctgccacccc gccagccttc cgccctggtg cccagtgtga 1320 ccgagtaccg cctggatggc cacaccatct cagacctgag ccggagcagc cggggcgagc 1380 tgatccccat ctcccccagc actgaagtcg ggggctctgg cattggcaca ccgccctctg 1440 tgctcaagcg gcagaggaag aggcgtgtgg ctctgtcccc tgtcactgag aatagcacca 1500 gtctgtcctt cctggattcc tgtaacagcc tcacgcccaa gagcacacct gttaagaccc 1560 tgcccttctc gccctcccag tttc tgaact tctggaacaa acaggacaca ttggagctgg 1620 agagcccctc gctgacatcc accccagtgt gcagccagaa ggtggtggtc accacaccac 1680 201200525 tgcaccggga caagacaccc ctgcaccaga aacatgctgc gtttgtaacc ccagatcaga 1740 agtactccat ggacaacact ccccacacgc caaccccgtt caagaacgcc ctggagaagt 1800 acggacccct gaagcccctg ccacagaccc cgcacctgga ggaggacttg aaggaggtgc 1860 tgcgttctga ggctggcatc gaactcatca tcgaggacga catcaggccc gagaagcaga 1920 agaggaagcc tgggctgcgg cggagcccca tcaagaaagt ccggaagtct ctggctcttg 1980 acattgtgga tgaggatgtg aagctgatga tgtccacact gcccaagtct ctatccttgc 2040 cgacaactgc cccttcaaac tcttccagcc tcaccctgtc aggtatcaaa gaagacaaca 2100 gcttgctcaa ccagggcttc ttgcaggcca agcccgagaa ggcagcagtg gcccagaagc 2160 cccgaagcca cttcacgaca cctgccccta tgtccagtgc ctggaagacg gtggcctgcg 2220 gggggaccag ggaccagctt ttcatgcagg agaaagcccg gcagctcctg ggccgcctga 2280 agcccagcca cacatctcgg accctcatct tgtcctgagg tgttgagggt gtcacgagcc 2340 cattctcatg tttacagggg ttgtgggggc agagggggtc tgtgaatctg agagtcattc 2400 aggtgacctc ctgcagggag ccttctgcca ccagcccctc cccagactct caggtggagg 2460 caacagggcc atgtgctgcc ctgttgccga gcccagctgt gggcggctcc tggtgctaac 2520 aacaaagttc cacttccagg tctgcctggt tccctcccca aggccacagg gagctccgtc 2580 agcttctccc aagcccacgt caggcctggc ctcatctcag accctgctta ggatggggga 2640 tgtggccagg ggtgctcctg tgctcaccct ctcttggtgc atttttttgg aagaataaaa 2700 ttgcctctct cttaaaaaaa aaaaaaaaaa a 2731 &lt; 210> 6 &lt; 211> 700 &lt; 212> PRT &lt;213>Human&lt;400&gt; 6 4 5 201200525

Met Ser Arg Arg Thr Arg Cys Glu Asp Leu Asp Glu Leu His Tyr Gin 15 10 15Met Ser Arg Arg Thr Arg Cys Glu Asp Leu Asp Glu Leu His Tyr Gin 15 10 15

Asp Thr Asp Ser Asp Val Pro Glu Gin Arg Asp Ser Lys Cys Lys Val 20 25 30Asp Thr Asp Ser Asp Val Pro Glu Gin Arg Asp Ser Lys Cys Lys Val 20 25 30

Lys Trp Thr His Glu Glu Asp Glu Gin Leu Arg Ala Leu Val Arg Gin 35 40 45Lys Trp Thr His Glu Glu Asp Glu Gin Leu Arg Ala Leu Val Arg Gin 35 40 45

Phe Gly Gin Gin Asp Trp Lys Phe Leu Ala Ser His Phe Pro Asn Arg 50 55 60Phe Gly Gin Gin Asp Trp Lys Phe Leu Ala Ser His Phe Pro Asn Arg 50 55 60

Thr Asp Gin Gin Cys Gin Tyr Arg Trp Leu Arg Val Leu Asn Pro Asp 65 70 75 80Thr Asp Gin Gin Cys Gin Tyr Arg Trp Leu Arg Val Leu Asn Pro Asp 65 70 75 80

Leu Val Lys Gly Pro Trp Thr Lys Glu Glu Asp Gin Lys Val Me Glu 85 90 95Leu Val Lys Gly Pro Trp Thr Lys Glu Glu Asp Gin Lys Val Me Glu 85 90 95

Leu Val Lys Lys Tyr Gly Thr Lys Gin Trp Thr Leu lie Ala Lys His 100 105 110Leu Val Lys Lys Tyr Gly Thr Lys Gin Trp Thr Leu lie Ala Lys His 100 105 110

Leu Lys Gly Arg Leu Gly Lys Gin Cys Arg Glu Arg Trp His Asn His 115 120 125Leu Lys Gly Arg Leu Gly Lys Gin Cys Arg Glu Arg Trp His Asn His 115 120 125

Leu Asn Pro Glu Val Lys Lys Ser Cys Trp Thr Glu Glu Glu Asp Arg 130 135 140 lie Me Cys Glu Ala His Lys Val Leu Gly Asn Arg Trp Ala Glu lie 145 150 155 160Leu Asn Pro Glu Val Lys Lys Ser Cys Trp Thr Glu Glu Glu Asp Arg 130 135 140 lie Me Cys Glu Ala His Lys Val Leu Gly Asn Arg Trp Ala Glu lie 145 150 155 160

Ala Lys Met Leu Pro Gly Arg Thr Asp Asn Ala Val Lys Asn His Trp 165 170 175 201200525Ala Lys Met Leu Pro Gly Arg Thr Asp Asn Ala Val Lys Asn His Trp 165 170 175 201200525

Asn Ser Thr Me Lys Arg Lys Val Asp Thr Gly Gly Phe Leu Ser Glu 180 185 190Asn Ser Thr Me Lys Arg Lys Val Asp Thr Gly Gly Phe Leu Ser Glu 180 185 190

Ser Lys Asp Cys Lys Pro Pro Val Tyr Leu Leu Leu Glu Leu Glu Asp 195 200 205Ser Lys Asp Cys Lys Pro Pro Val Tyr Leu Leu Leu Glu Leu Glu Asp 195 200 205

Lys Asp Gly Leu Gin Ser Ala Gin Pro Thr Glu Gly Gin Gly Ser Leu 210 215 220Lys Asp Gly Leu Gin Ser Ala Gin Pro Thr Glu Gly Gin Gly Ser Leu 210 215 220

Leu Thr Asn Trp Pro Ser Val Pro Pro Thr lie Lys Glu Glu Glu Asn 225 230 235 240Leu Thr Asn Trp Pro Ser Val Pro Pro Thr lie Lys Glu Glu Glu Asn 225 230 235 240

Ser Glu Glu Glu Leu Ala Ala Ala Thr Thr Ser Lys Glu Gin Glu Pro 245 250 255 lie Gly Thr Asp Leu Asp Ala Val Arg Thr Pro Glu Pro Leu Glu Glu 260 265 270Ser Glu Glu Glu Leu Ala Ala Ala Thr Thr Ser Lys Glu Gin Glu Pro 245 250 255 lie Gly Thr Asp Leu Asp Ala Val Arg Thr Pro Glu Pro Leu Glu Glu 260 265 270

Phe Pro Lys Arg Glu Asp Gin Glu Gly Ser Pro Pro Glu Thr Ser Leu 275 280 285Phe Pro Lys Arg Glu Asp Gin Glu Gly Ser Pro Pro Glu Thr Ser Leu 275 280 285

Pro Tyr Lys Trp Val Val Glu Ala Ala Asn Leu Leu lie Pro Ala Val 290 295 300Pro Tyr Lys Trp Val Val Glu Ala Ala Asn Leu Leu lie Pro Ala Val 290 295 300

Gly Ser Ser Leu Ser Glu Ala Leu Asp Leu lie Glu Ser Asp Pro Asp 305 310 315 320Gly Ser Ser Leu Ser Glu Ala Leu Asp Leu lie Glu Ser Asp Pro Asp 305 310 315 320

Ala Trp Cys Asp Leu Ser Lys Phe Asp Leu Pro Glu Glu Pro Ser Ala 325 330 335Ala Trp Cys Asp Leu Ser Lys Phe Asp Leu Pro Glu Glu Pro Ser Ala 325 330 335

Glu Asp Ser lie Asn Asn Ser Leu Val Gin Leu Gin Ala Ser His Gin 340 345 350 6 201200525Glu Asp Ser lie Asn Asn Ser Leu Val Gin Leu Gin Ala Ser His Gin 340 345 350 6 201200525

Gin Gin Val Leu Pro Pro Arg Gin Pro Ser Ala Leu Val Pro Ser Val 355 360 365Gin Gin Val Leu Pro Pro Arg Gin Pro Ser Ala Leu Val Pro Ser Val 355 360 365

Thr Glu Tyr Arg Leu Asp Gly His Thr Me Ser Asp Leu Ser Arg Ser 370 375 380Thr Glu Tyr Arg Leu Asp Gly His Thr Me Ser Asp Leu Ser Arg Ser 370 375 380

Ser Arg Gly Glu Leu lie Pro lie Ser Pro Ser Thr Glu Val Gly Gly 385 390 395 400Ser Arg Gly Glu Leu lie Pro lie Ser Pro Ser Thr Glu Val Gly Gly 385 390 395 400

Ser Gly Me Gly Thr Pro Pro Ser Val Leu Lys Arg Gin Arg Lys Arg 405 410 415Ser Gly Me Gly Thr Pro Pro Ser Val Leu Lys Arg Gin Arg Lys Arg 405 410 415

Arg Val Ala Leu Ser Pro Val Thr Glu Asn Ser Thr Ser Leu Ser Phe 420 425 430Arg Val Ala Leu Ser Pro Val Thr Glu Asn Ser Thr Ser Leu Ser Phe 420 425 430

Leu Asp Ser Cys Asn Ser Leu Thr Pro Lys Ser Thr Pro Val Lys Thr 435 440 445Leu Asp Ser Cys Asn Ser Leu Thr Pro Lys Ser Thr Pro Val Lys Thr 435 440 445

Leu Pro Phe Ser Pro Ser Gin Phe Leu Asn Phe Trp Asn Lys Gin Asp 450 455 460Leu Pro Phe Ser Pro Ser Gin Phe Leu Asn Phe Trp Asn Lys Gin Asp 450 455 460

Thr Leu Glu Leu Glu Ser Pro Ser Leu Thr Ser Thr Pro Val Cys Ser 465 470 475 480Thr Leu Glu Leu Glu Ser Pro Ser Leu Thr Ser Thr Pro Val Cys Ser 465 470 475 480

Gin Lys Val Val Val Thr Thr Pro Leu His Arg Asp Lys Thr Pro Leu 485 490 495Gin Lys Val Val Val Thr Thr Pro Leu His Arg Asp Lys Thr Pro Leu 485 490 495

His Gin Lys His Ala Ala Phe Val Thr Pro Asp Gin Lys Tyr Ser Met 500 505 510His Gin Lys His Ala Ala Phe Val Thr Pro Asp Gin Lys Tyr Ser Met 500 505 510

Asp Asn Thr Pro His Thr Pro Thr Pro Phe Lys Asn Ala Leu Glu Lys 515 520 525 201200525Asp Asn Thr Pro His Thr Pro Thr Pro Phe Lys Asn Ala Leu Glu Lys 515 520 525 201200525

Tyr Gly Pro Leu Lys Pro Leu Pro Gin Thr Pro His Leu Glu Glu Asp 530 535 540Tyr Gly Pro Leu Lys Pro Leu Pro Gin Thr Pro His Leu Glu Glu Asp 530 535 540

Leu Lys Glu Val Leu Arg Ser Glu Ala Gly lie Glu Leu lie Me Glu 545 550 555 560Leu Lys Glu Val Leu Arg Ser Glu Ala Gly lie Glu Leu lie Me Glu 545 550 555 560

Asp Asp I le Arg Pro Glu Lys Gin Lys Arg Lys Pro Gly Leu Arg Arg 565 570 575Asp Asp I le Arg Pro Glu Lys Gin Lys Arg Lys Pro Gly Leu Arg Arg 565 570 575

Ser Pro lie Lys Lys Val Arg Lys Ser Leu Ala Leu Asp Me Val Asp 580 585 590Ser Pro lie Lys Lys Val Arg Lys Ser Leu Ala Leu Asp Me Val Asp 580 585 590

Glu Asp Val Lys Leu Met Met Ser Thr Leu Pro Lys Ser Leu Ser Leu 595 600 605Glu Asp Val Lys Leu Met Met Ser Thr Leu Pro Lys Ser Leu Ser Leu 595 600 605

Pro Thr Thr Ala Pro Ser Asn Ser Ser Ser Leu Thr Leu Ser Gly lie 610 615 620Pro Thr Thr Ala Pro Ser Asn Ser Ser Ser Leu Thr Leu Ser Gly lie 610 615 620

Lys Glu Asp Asn Ser Leu Leu Asn Gin Gly Phe Leu Gin Ala Lys Pro 625 630 635 640Lys Glu Asp Asn Ser Leu Leu Asn Gin Gly Phe Leu Gin Ala Lys Pro 625 630 635 640

Glu Lys Ala Ala Val Ala Gin Lys Pro Arg Ser His Phe Thr Thr Pro 645 650 655Glu Lys Ala Ala Val Ala Gin Lys Pro Arg Ser His Phe Thr Thr Pro 645 650 655

Ala Pro Met Ser Ser Ala Trp Lys Thr Val Ala Cys Gly Gly Thr Arg 660 665 670Ala Pro Met Ser Ser Ala Trp Lys Thr Val Ala Cys Gly Gly Thr Arg 660 665 670

Asp Gin Leu Phe Met Gin Glu Lys Ala Arg Gin Leu Leu Gly Arg Leu 675 680 685Asp Gin Leu Phe Met Gin Glu Lys Ala Arg Gin Leu Leu Gly Arg Leu 675 680 685

Lys Pro Ser His Thr Ser Arg Thr Leu lie Leu Ser 690 695 700 ⑤ 201200525 &lt;210&gt; 7 &lt;211〉 22 &lt;212〉 DNA 〈213&gt;人工 &lt;220&gt; &lt;223〉人工序列 &lt;400〉 7 gtctaccagg cattcgcttc at 22 241 人 &lt;210&gt; &lt;211〉 〈212&gt; &lt;213〉 &lt;220&gt; 〈223&gt;人工序列 &lt;400〉 8 tcagctggac cacagccgca gcgt 24 &lt;210〉 211 人 &lt;211〉 &lt;212〉 &lt;213〉 &lt;220〉 &lt;223〉人工序列 &lt;400〉 9 tcagaaatcc tttctcttga c 21 &lt;210&gt; 10 &lt;211〉 24 &lt;212&gt; DNA 〈213&gt;人工 &lt;220&gt; &lt;223〉人工序列 9 201200525 &lt;400〉 10 ctagcctctg gaatcctttc tctt 24Lys Pro Ser His Thr Ser Arg Thr Leu lie Leu Ser 690 695 700 5 201200525 &lt;210&gt; 7 &lt;211> 22 &lt;212> DNA <213> Manual&lt;220&gt;&lt;223>ArtificialSequence&lt;400&gt; 7 gtctaccagg cattcgcttc at 22 241 person &lt;210&gt;&lt;211> <212> &lt;213> &lt;220&gt;<223>&gt; artificial sequence&lt;400> 8 tcagctggac cacagccgca gcgt 24 &lt;210> 211 person&lt;211> &lt;212&gt;&lt;213&gt;&lt;220&gt;&lt;223&gt; artificial sequence&lt;400&gt; 9 tcagaaatcc tttctcttga c 21 &lt;210&gt; 10 &lt;211> 24 &lt;212&gt; DNA <213>&lt;220&gt;&lt;223>Artificial sequence 9 201200525 &lt;400> 10 ctagcctctg gaatcctttc tctt 24

Claims (1)

201200525 七、申請專利範圍: 1. 一種經分離的募胜肽,包括一胺基酸序列,其係擇 自由序列辨識號:2至4所組成之群組,其中該寡胜肽與 一人類白血球組織抗原結合且具有細胞毒殺性T淋巴球誘 發能力。 2·—種經分離的寡胜肽,其中該寡胜肽與一人類白血 球組織抗原結合且具有細胞毒殺性T淋巴球誘發能力,其 中該募胜肽係由擇自序列辨識號:2至4中之一胺基酸序 列所組成,其中1、2或數個胺基酸被插入、取代、刪除或 加入。 3_如申請專利範圍第2項所述之募胜肽,其中該寡胜 肽具有下列特徵之一或兩者: (a) 來自N端之第二個胺基酸係擇自白胺酸與曱硫丁 胺酸中;以及 (b) C端胺基酸係擇自纈氨酸與白胺酸中。 4.如申清專利祀圍第1至3項所述之經分離的寡胜 肽’其中該人類白血球組織抗原為人類白血球組織抗原 _A2 〇 5·如申晴專利範圍第1至4項所述之經分離的寡胜 肽’其中該胜肽為九胜肽或十胜肽。 6. 一種經分離之多核苦酸’其編碼出如申請專利範圍 第1至5項之任一項所述的寡胜肽。 7. —種誘發細胞毒殺性τ淋巴球之試劑,其中該試劑 包括如申請專利範圍第丨至5項之任一項所述之一或多個 201200525 該寡胜狀,或如申古杳蛮丄丨 甲叫專利範圍第6項所述之一或多個該多 核苦酸。 8· 一種藥學試劑,用於癌症之治療及/或預防,及/或 八手術後復發的避免’纟中該試劑包括如中請專利範圍第 1至5項之任項所述之一或多個該募胜肽,或如申請專 利範圍第6項所述之—或多個該多核苦酸。 9. 如申請專利範圍第8項所述之藥學試劑被配製來 用以投予一個體,其人類白血球組織抗原為人類白血球組 織抗原-A2。 10. 如申請專利範圍第8或9項所述之藥學試劑,其被 配製來用於治療癌症。 11. 一種誘導具有細胞毒殺性τ淋巴球誘發能力之抗 原呈現細胞的方法,其中該方法包括下列步驟之一: (a) i/3 la tr0、h叩或y刀h叩將一抗原呈現細 胞與如申請專利範圍第1至5項之任一項所述之募胜肽接 觸;或 (b) 將編碼出如申請專利範圍第1至5項之任一項所 述之寡胜肽的一多核苷酸引入一抗原呈現細胞。 12· 一種誘導細胞毒殺性T淋巴球的方法,藉由任何 之包括下列步驟之至少一個的方法: (a) 將CD8陽性τ細胞與抗原呈現細胞共培養,抗原 呈現細胞表現一人類白血球組織抗原與如申請專利範圍第 1至5項之任一項所述之寡胜肽的一複合物於其表面上; (b) 將CD8陽性τ細胞與外吐小體共培養,外吐小體 201200525 表現一人類白血球組織抗原與如申請專利範圍第1至5項 之任一項所述之募胜肽的一複合物於其表面上;以及 (c)將一包括編碼出一 τ細胞受體次單元多胜肽之多 核普酸的基因引入一 T細胞,該τ細胞受體次單元多胜肽 與如申請專利範圍第1至5項之任一項所述的寡胜肽結合。 1 3 · —種經分離之抗原呈現細胞,其表現一人類白血球 組織抗原與如申請專利範圍第1至5項之任一項所述之寡 胜肽的一複合物於其表面上。 14.如申請專利範圍第13項所述之抗原呈現細胞,其 藉由如申請專利範圍第11項所述之方法來誘導。 1 5. —種經分離之細胞毒殺性T淋巴球,其以如申請專 利範圍第1至5項之任一項所述之胜肽為標的。 16. 如申請專利範圍第丨5項所述之細胞毒殺性τ淋巴 球’其藉由如申請專利範圍第12項所述之方法來誘導。 17. —種於一個體中誘導抗癌症之免疫反應的方法,其 包括下列步驟:投予該個體一試劑’該試劑包括如申請專 利範圍第1至5項之任一項所述之寡胜肽、一其免疫活性 片段,或一編碼出該募胜肽或免疫活性片段之一多核苷酸。201200525 VII. Patent application scope: 1. An isolated recruit peptide, including an amino acid sequence, which is selected from the group consisting of 2 to 4, wherein the oligopeptide and a human leukocyte Tissue antigen binds and has a cytotoxic T lymphocyte evoking ability. 2. An isolated oligopeptide, wherein the oligopeptide binds to a human leukocyte antigen and has a cytotoxic T lymphocyte evoking ability, wherein the peptide is selected from sequence identification numbers: 2 to 4. One of the amino acid sequences consisting of 1, 2 or several amino acids inserted, substituted, deleted or added. 3) The peptide as described in claim 2, wherein the oligopeptide has one or both of the following characteristics: (a) the second amino acid from the N-terminus is selected from leucine and hydrazine In thiobutyric acid; and (b) the C-terminal amino acid is selected from the group consisting of proline and leucine. 4. The isolated oligopeptide described in paragraphs 1 to 3 of the patent application, wherein the human leukocyte antigen is a human leukocyte antigen _A2 〇5, such as Shen Qing patent scope items 1 to 4 The isolated oligopeptide is described as wherein the peptide is a nine-peptide or a ten-peptide. An isolated polynucleic acid, which encodes the oligopeptide according to any one of claims 1 to 5. 7. An agent for inducing a cytotoxic thymocyte, wherein the reagent comprises one or more of 201200525 as described in any one of claims 5 to 5, or such as Shen Guzhen The armor is called one or more of the polynucleic acid as described in item 6 of the patent scope. 8. A pharmaceutical agent for the treatment and/or prevention of cancer, and/or avoidance of recurrence after eight operations, wherein the agent comprises one or more of any one of items 1 to 5 of the patent scope of the patent application. The recruiting peptide, or as described in claim 6 of the patent application, or a plurality of the polynucleic acid. 9. The pharmaceutical agent according to item 8 of the patent application is formulated for administration to a human, and the human leukocyte tissue antigen is human leukocyte tissue antigen-A2. 10. The pharmaceutical agent of claim 8 or 9, which is formulated for the treatment of cancer. A method for inducing an antigen-presenting cell having a cytotoxic lymphoblast-inducing ability, wherein the method comprises one of the following steps: (a) i/3 la tr0, h叩 or y knife h叩 presenting an antigen to a cell Contacting the acetonide as described in any one of claims 1 to 5; or (b) encoding one of the oligopeptides as described in any one of claims 1 to 5 The polynucleotide introduces an antigen presenting cell. 12. A method of inducing cytotoxic T lymphocytes by any method comprising at least one of the following steps: (a) co-cultivating CD8-positive tau cells with antigen-presenting cells, the antigen-presenting cells exhibiting a human leukocyte antigen a complex of the oligopeptide according to any one of claims 1 to 5 on the surface thereof; (b) co-culturing the CD8-positive tau cells with the exosome, exosome 201200525 Demonstrating a human leukocyte tissue antigen on a surface thereof with a complex of the peptide as described in any one of claims 1 to 5; and (c) encoding a tau cell receptor The multi-nucleotide gene of the unit polypeptide is introduced into a T cell which binds to the oligopeptide described in any one of claims 1 to 5. An isolated antigen-presenting cell exhibiting a complex of a human leukocyte antigen and a oligopeptide as described in any one of claims 1 to 5 on the surface thereof. 14. The antigen-presenting cell of claim 13, which is induced by the method of claim 11 of the patent application. An isolated cytotoxic T lymphocyte, which is characterized by a peptide as described in any one of claims 1 to 5. 16. The cytotoxic τ lymphocyte as described in claim 5, which is induced by the method of claim 12 of the patent application. 17. A method of inducing an immune response against cancer in a body, comprising the steps of: administering to the individual an agent comprising: the agent according to any one of claims 1 to 5; A peptide, an immunologically active fragment thereof, or a polynucleotide encoding one of the recruit peptide or the immunologically active fragment.
TW099141433A 2009-12-04 2010-11-30 MYBL2 peptides and vaccines containing the same TW201200525A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US26687109P 2009-12-04 2009-12-04

Publications (1)

Publication Number Publication Date
TW201200525A true TW201200525A (en) 2012-01-01

Family

ID=44114795

Family Applications (1)

Application Number Title Priority Date Filing Date
TW099141433A TW201200525A (en) 2009-12-04 2010-11-30 MYBL2 peptides and vaccines containing the same

Country Status (14)

Country Link
US (1) US20120328638A1 (en)
EP (1) EP2507257A4 (en)
JP (1) JP2013512659A (en)
KR (1) KR20120114284A (en)
CN (1) CN102753567A (en)
AU (1) AU2010327891A1 (en)
BR (1) BR112012013371A2 (en)
CA (1) CA2782484A1 (en)
IL (1) IL219927A0 (en)
MX (1) MX2012006376A (en)
RU (1) RU2012127762A (en)
SG (1) SG181108A1 (en)
TW (1) TW201200525A (en)
WO (1) WO2011067933A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108623657B (en) * 2018-05-11 2021-02-12 苏州大学 Polypeptide, recombinant DNA molecule, recombinant vector, exosome and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2375299T3 (en) * 2002-09-12 2012-02-28 Oncotherapy Science, Inc. KDR PEPTIDES AND VACCINES THAT CONTAIN THEM.
CN100352842C (en) * 2002-10-31 2007-12-05 詹森药业有限公司 Genes whose expression is increased in response to stimulation by corticotropin-releasing hormone
DE602006021283D1 (en) * 2005-02-25 2011-05-26 Oncotherapy Science Inc PEPTIDE VACCINE FOR LUNG CANCER EXPRESSING TTK POLYPEPTIDE
TW200908998A (en) * 2007-06-27 2009-03-01 Oncotherapy Science Inc Compositions and methods of treating cancer
TW201000119A (en) * 2008-06-10 2010-01-01 Oncotherapy Science Inc MYBL2 epitope peptides and vaccines containing the same

Also Published As

Publication number Publication date
SG181108A1 (en) 2012-07-30
KR20120114284A (en) 2012-10-16
IL219927A0 (en) 2012-07-31
MX2012006376A (en) 2012-07-10
CN102753567A (en) 2012-10-24
BR112012013371A2 (en) 2016-12-13
AU2010327891A1 (en) 2012-06-21
US20120328638A1 (en) 2012-12-27
CA2782484A1 (en) 2011-06-09
WO2011067933A1 (en) 2011-06-09
EP2507257A4 (en) 2013-04-03
EP2507257A1 (en) 2012-10-10
JP2013512659A (en) 2013-04-18
RU2012127762A (en) 2014-01-10

Similar Documents

Publication Publication Date Title
JP5799394B2 (en) NEIL3 peptide and vaccine containing the same
TW201138806A (en) CDCA5 peptides and vaccines including the same
TWI485245B (en) Modified melk peptides and vaccines containing the same
US9585948B2 (en) TOMM34 peptides and vaccines including the same
TW201414752A (en) UBE2T peptides and vaccines containing the same
TW201302800A (en) SEMA5B peptides and vaccines including the same
RU2531348C2 (en) Ttk peptides and vaccines containing them
TW201043244A (en) VANGL1 peptides and vaccines including the same
KR101863557B1 (en) Ect2 peptides and vaccines including the same
JP5838482B2 (en) HJURP peptide and vaccine containing the same
TW201106962A (en) CDC45L peptides and vaccines including the same
US8697631B2 (en) TMEM22 peptides and vaccines including the same
TW201100090A (en) C6orf167 peptides and vaccines containing the same
US20150322112A1 (en) Sema5b peptides and vaccines containing the same
TW201200525A (en) MYBL2 peptides and vaccines containing the same
TW201216982A (en) WDHD1 peptides and vaccines including the same
TW201211250A (en) VANGL1 peptides and vaccines including the same
TW201134480A (en) CLUAP1 peptides and vaccines including the same